You are on page 1of 15

Received: 29 July 2022 Revised: 3 March 2023 Accepted: 19 March 2023

DOI: 10.1111/ejn.15971

RESEARCH REPORT

A novel Reelin construct, R36, recovered behavioural


deficits in the heterozygous reeler mouse

Nicole K. Morrill | Qingyou Li | Aurelie Joly-Amado | Edwin J. Weeber |


Kevin R. Nash

Department of Molecular Pharmacology


and Physiology, Morsani College of Abstract
Medicine, University of South Florida, Reelin, a large extracellular glycoprotein, plays a critical role in prenatal brain
Tampa, FL, USA
development and postnatally in synaptic plasticity, learning and memory. Dys-
Correspondence regulation of Reelin signalling has been implicated in several neuropsychiatric
Kevin R. Nash, Department of Molecular disorders including schizophrenia, autism, depression and Alzheimer’s dis-
Pharmacology and Physiology, Morsani
College of Medicine, University of South
ease. Previous studies have demonstrated that Reelin’s central fragment,
Florida, 12901 Bruce B. Downs Blvd., R3456, binds to ApoER2, inducing ApoER2 clustering and subsequent intra-
Tampa, FL 33612, USA. cellular signalling. We previously reported the development of a novel lucifer-
Email: nash@usf.edu
ase complementation assay, which we used to demonstrate that R3456 can
Edited by: Christina Dalla lead to ApoER2 receptor dimerization. Using this same assay, we explored var-
ious smaller fragments and combinations from R3456, and we identified a con-
struct of repeats 3 and 6 (R36), which could still elicit equivalent receptor
dimerization. The purpose of this study was to test R36 for biological effects
in vitro and in vivo. We show that R36 was capable of initiating intracellular
signalling in primary neuronal cultures. In addition, we demonstrate that a
single intracerebroventricular injection of R36 protein into a model of Reelin
deficiency, the heterozygous reeler mice, can significantly improve cognition.
These data support a role for the new construct R36 to enhance the Reelin
pathway, and the future possibility of exploring gene therapy approaches with
R36 in diseases characterized by reduced levels of Reelin.

KEYWORDS
HRM model, Reelin, Reelin supplementation, therapeutics

1 | INTRODUCTION
Abbreviations: AKT, protein kinase B; AMPA, α-amino-3-hydroxy-
Reelin is a large glycoprotein (450 kDa) and contains an
5-methyl-4-isoxazolepropionic acid; ApoER2, apolipoproten E receptor
2; CDK, cyclin-dependent kinase; Dab1, Disabled-1; EGF, Epidermal F-spondin homology region located at the N-terminus,
growth factor; ERK, extracellular signal-regulated kinase; GABA, eight repeats and a small positively charged C-terminus
Gamma-aminobutyric acid; GSK3β, glycogen synthase kinase 3-beta; (Figure 1a) (D’Arcangelo et al., 1995, 1997). Each repeat
HRM, heterozygous reeler mouse; ICV, intracerebroventricular; NMDA, contains two subdomains A and B, separated by an epi-
N-methyl-D-aspartate; VLDLR, very low density lipoprotein receptor. dermal growth factor (EGF)-like motif (D’Arcangelo
Nicole K. Morrill and Qingyou Li are co-first authors. et al., 1997; Jossin et al., 2004; Royaux et al., 1997). Reelin
© 2023 Federation of European Neuroscience Societies and John Wiley & Sons Ltd.

Eur J Neurosci. 2023;57:1657–1670. wileyonlinelibrary.com/journal/ejn 1657


14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1658 MORRILL ET AL.

F I G U R E 1 Reelin fragments R3456 and R36 increase firefly luciferase activity. (a) Reelin is composed of eight EGF-like repeats and
undergoes cleavage at two sites as notated with black arrows, between repeats 2 and 3 and repeats 6 and 7, resulting in five naturally
occurring fragments as depicted above. This project focuses on naturally occurring repeat, R3456 and a novel made repeat, R36.
(b) HEK293T cells were co-transfected with pΔEx5-NFluc and pΔEx5-CFluc with or without pSec-R3456 or pSec-R36. Firefly luciferase
activity was increased by R3456 and R36 when compared with the ApoER2-Luc construct, F3,12 = 2.886. Data shown as mean ± SEM. For
all groups, n = 4, **p < 0.01, ****p < 0.0001.

undergoes natural cleavage at two sites, between repeats


memory. Reelin is involved in potentiating glutamatergic
2 and 3 and repeats 6 and 7, generating five naturally
and GABAergic neurotransmission, aids in synaptic mat-
occurring fragments (Ignatova et al., 2004; Lambert de
uration and increases AMPA and NMDA receptor expres-
Rouvroit et al., 1999). Reelin can bind and signal through
sion and activity (reviewed (Jossin, 2020; Lee &
two known receptors, apolipoprotein E receptor
D’Arcangelo, 2016). Reductions in Reelin levels and sig-
2 (ApoER2) and very low density lipoprotein receptor
(VLDLR) (Beffert et al., 2004, 2006; D’Arcangelo nalling have been associated with a number of neurologi-
et al., 1999; Hiesberger et al., 1999; Qiu et al., 2006). Both cal diseases including schizophrenia, autism, temporal
ApoER2 and VLDLR bind to the ligands apolipoprotein lobe epilepsy and Alzheimer’s disease (Cuchillo-Ibanez
E (ApoE) and Reelin, which can compete for receptor et al., 2016; Fatemi et al., 2005; Folsom & Fatemi, 2013;
binding (D’Arcangelo et al., 1999; Hiesberger Ishii et al., 2016; Jossin, 2020). In humans, loss of Reelin
et al., 1999). It has been previously demonstrated with results in a type of lissencephaly with severe cortical and
immunoprecipitation experiments that the central frag- cerebellar malformation (Hong et al., 2000). Thus, there
ment containing repeats 3 through 6 or R3456 (190 kDa) is a growing body of interest in finding potential thera-
is the fragment involved in initiating the Reelin signal- peutic strategies that target Reelin signalling and supple-
ling cascade and interacts with VLDLR and ApoER2 mentation. The mouse reln gene spans 450 kb contains
(Jossin et al., 2004; Royaux et al., 1997). Receptor cluster- 65 exons and was first characterized in the homozygous
ing leads to the recruitment and tyrosine phosphorylation reeler mouse model. Anatomically, homozygous reeler
of Disabled-1 (Dab1), via Src family kinases (Howell mice are characterized by abnormalities in neuronal layer
et al., 1999, 2000; Strasser et al., 2004). Phosphorylation formation and positioning in brain regions during devel-
of Dab1 leads to activation of multiple downstream sig- opment, leading to malformations of neocortex, cerebel-
nalling pathways. These pathways include activation of lum and hippocampus (Boyle et al., 2011;
the Src-family of tyrosine kinases (Arnaud et al., 2003; Hamburgh, 1963; Tissir & Goffinet, 2003). These mice
Ballif et al., 2003; Jossin et al., 2003), AKT (Ballif exhibit dystonic posture, tremors and a dramatic ataxic
et al., 2003), PI3K (Bock et al., 2003; Jossin & gait, known as “reeling” phenotype as early as 2 weeks of
Goffinet, 2007), ERK (Lee et al., 2014) pathways and the age, together with enhanced susceptibility to seizure
CDK pathway (Ohkubo et al., 2003; Ohshima et al., 2001; (Curran & D’Arcangelo, 1998; Falconer, 1951; Patrylo
Yamamoto et al., 2009; Yoneshima et al., 1997). This acti- et al., 2006; Tissir & Goffinet, 2003). Moreover, viability,
vation can lead to effects on cell adhesion, dendrite out- fertility and immunity are greatly reduced in these mice
growth and spine formation, as well as synaptic receptor (Green-Johnson et al., 1995; Meseke et al., 2018; Togo
reorganization. et al., 1986). These severe deficits render difficult the use
In the developing brain, Reelin plays a critical role in of this model for testing of potential therapeutic targets.
guiding radial migration of neurons, whereas in adult- The heterozygous reeler mouse (HRM) model is produced
hood Reelin supports synaptic plasticity and learning and through the heterozygous Reelin mutation resulting in a
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MORRILL ET AL. 1659

50% reduction of the Reelin protein in the CNS through- Male and female mice aged 12–14 weeks were used
out development and the life of the animal. Although less within this study and the groups were as follows: HRM
severe than the homozygous reeler mice, the HRM show given the PBS injection (5M, 5F), HRM given the R36
biochemical alterations in spine number and morphol- protein injection (5M, 5F), WT mice given the PBS injec-
ogy, and a distinct defect in hippocampal synaptic plastic- tion (6M, 4F) and WT mice given the R36 protein injec-
ity and memory dysfunction (Barr et al., 2008; Qiu tion (6M, 4F).
et al., 2006). Furthermore, treatment of this model with
Reelin recovered the biochemical, morphological and
physiological deficits in the adult HRM mouse. Even 2.3 | Constructs and vector
more compelling was the recovery of HRM-related beha-
vioural and cognitive impairments (Rogers et al., 2013). Central fragment of human Reelin encoding amino acids
We have previously presented the development of a 1313 to 2618 of Reelin was purified as previously
novel luciferase complementation assay, which we used described (Li et al., 2023). The R36 construct was synthe-
to demonstrate that R3456 can lead to receptor dimeriza- sized by Gen9 Inc. (Cambridge, MA) and encompassed
tion or clustering (Li et al., 2023). Here, we used this fire- the human Reelin amino acid fragments 1313–1614
fly luciferase assay to identify smaller fragments of Reelin (R3) and 2329–2618 (R6). R36 was ligated into the pSec-
which are capable of inducing receptor clustering. We Tag2B vector in frame with the Ig-kappa leader sequence
present a new fragment of Reelin that can enhance and the His- and Myc-tags, similar to R3456 as described
Reelin pathway in vitro. We then test Reelin enhance- previously (Li et al., 2023).
ment through our novel Reelin fragment in the heterozy-
gous reeler mice. This study aids in supporting the
possibility of Reelin supplementation as a therapeutic 2.4 | Production and purification of R36
and raises the need for further work in other models
affected by a loss of Reelin expression. A stably transfected HEK293T cell line expressing R36
was achieved with the selection reagent Zeocin
(InvivoGen) and Dot blots to screen for positive clones.
2 | MATERIALS AND METHODS The cells were cultured in Dulbecco’s modified Eagle’s
medium (DMEM) containing 10% foetal bovine serum
2.1 | Materials (FBS) and 100-μg/mL Zeocin (InvivoGen) at 37 C and 5%
CO2. Reelin was purified from the conditioned medium
The materials used are as follows: Zeocin (InvivoGen, using a Ni-Sepharose™ Excel resin (GE) column as
ant-zn); anti-Dab1 (Millipore, AB5840); anti-pDab1 described in detail previously (Li et al., 2023).
(Millipore, AB9644); anti-β Actin (Cell Signaling 4967S);
RIPA Lysis buffer (Millipore, Tris/HCl pH 7.4, 1 mM
EDTA, 150 mM NaCl, 1% NP40, 0.25% sodium deoxycho- 2.5 | Cell culture
late). The following were obtained from Cell Signaling:
Anti-pERK, anti-ERK; anti-Akt; anti-pAkt. For luciferase assay and transient transfections,
HEK293T cells were grown and maintained in DMEM
(Sigma-Aldrich) with 10% FBS and 1% penicillin/
2.2 | Animal handling streptomycin at 37 C and 5% CO2. For primary neurons,
cortical tissue including the hippocampus was isolated on
Wide-type C57BL/6J (WT) mice were obtained from embryonic day 16 (E16) from C57BL/6 mouse pups. The
Jackson laboratories and were bred for this study in minced tissues were gently dissociated using 0.25% tryp-
accordance with IACUC breeding protocols (Bar Harbor, sin and mechanical trituration to achieve individual cor-
Maine, USA). Reeler heterozygous mice (HRM) (B6C3Fe tical neuronal cells. The neuronal cells were plated on a
a/a-Relnrl/J; Jackson Laboratories) were generated by six-well plate coated with 0.1% poly-L-lysine (Sigma-
heterozygous crosses and were bred for this study in Aldrich) at a density of 200,000 cells/well and incubated
accordance with IACUC breeding protocols. Animals in 3-mL of neurobasal medium (Gibco) supplemented
were housed in standard 12-h light–dark cycle with with 2% B-27 (Invitrogen) and 0.5-mM GlutaMax (Gibco)
access to food and water ad libitum. All animal care pro- at 37 C and 5% CO2. On day in vitro (DIV) 14, the
tocols used were approved by the Institutional Animal medium was replaced with neurobasal medium contain-
Care and Use Committee of the University of South Flor- ing various concentrations of R36 protein (0.2 to 1 nM)
ida (PHS Assurance number: D-16-00589 [A4100-01]). for 1 h at 37 C and 5% CO2.
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1660 MORRILL ET AL.

2.6 | Luciferase assay 20 prior to incubation with the appropriate secondary


antibody with HRP-conjugation (GE, anti-mouse IgG and
Dual-Glo luciferase assay was performed in HEK293T anti-rabbit IgG 1:3000) for 1 h at room temperature. Fol-
cells as previously described in detail in Li et al. (2023). lowing a repeated washing, the blots were developed with
Cells were co-transfected with pΔEx5-NFluc and an enhanced chemiluminescence reagent
pΔEx5-CFluc with or without pSec-R3456 or pSec-R36 to (ThermoFisher), then exposed to an X-ray film which
express firefly luciferase. HEK293T cells were plated onto was quantified by using Image J software (NIH).
a 24-well-plate at a density of 2  105 cells/well in Western Blot analysis of pre- and post-synaptic
DMEM media supplemented with 10% foetal bovine markers: Dissected mouse brain regions were homoge-
serum and 1% penicillin/streptomycin, and the cultures nized in RIPA buffer for western blot analysis as detailed
were incubated at 37 C and 5% CO2 overnight. The fol- above. We used the following primary antibodies: anti-
lowing day, the medium was replaced by Opti-MEM post-synaptic density protein-95 (PSD95) antibody
serum-free medium, and the constructs were co- (Abcam; 1:500) and anti-synaptophysin antibody
transfected with Lipofectamine 2000 (ThermoFisher). (Abcam; 1:1000). Revert total protein (LI-COR) was used
Approximately 48 h post-transfection, the medium was as a protein control. Membranes were washed for 10 min,
removed, and Glo Lysis Buffer (Promega) was added onto 3X in TBS with 0.1% Tween 20 followed by incubation
the plate to lyse the cells for 5 min. The cells were then with fluorescently tagged secondary antibody (anti-
centrifuged, and the supernatant and Dual-Glo Lucifer- mouse IgG and anti-rabbit IgG, 1:10,000, respectively) for
ase assay reagent combined in a 96-well assay plate. The 1 h. Proceeding washing, the blots were scanned and
firefly luminescence was first measured by using a Bio- quantified using an Odyssey Dlx (LI-COR biosciences)
Tek Synergy H4 plate reader. scanner and software.

2.7 | Western-blot analysis 2.8 | Direct bilateral ventricular


injections
HEK293T cells were lysed in Glo Lysis Buffer and cortical
neurons were lysed in RIPA Lysis buffer (Millipore, Tris/ Mice were anaesthetized with isoflurane and placed on a
HCl pH 7.4, 1-mM EDTA, 150-mM NaCl, 1% NP40, stereotaxic surgery apparatus (World Precision Instru-
0.25% sodium deoxycholate) containing complete prote- ments). A subcutaneous injection of Nocita (bupivacaine
ase inhibitors (Sigma) and phosphatase inhibitors I and liposome injectable suspension, Elanco) 20 μL at
II (Sigma) and then centrifuged at 13,000 g for 12 min at 13.3 mg/mL was performed on the head of the animal as
4 C. The supernatants were collected, and the protein a long-lasting post-surgical pain analgesic. After 2 min, a
concentrations were measured using a standard BCA sagittal incision was made mid-cranium and the skin
assay (Pierce, Rockford, IL). Samples were prepared in gently pushed back to enlarge the opening. Two holes
Laemmli’s sample buffer (Bio-Rad) with 5% were drilled through the skull to allow passage of the
2-mercaptoethanol and heated for 95 C for 10-min. Pro- Hamilton needle through the brain, into the ventricles
teins were run on 4–15% TGX Criterion gels (Bio-Rad). (AP 0.4 mm, L ±1.0 mm, and V 2.4 mm from
Gels were transferred to Trans-Blot® Turbo™ Midi Nitro- bregma). Mice were injected bilaterally with 3.0-μL PBS
cellulose blots, followed by blocking with 5% nonfat dry or 3.0 μL of Reelin (R36) at 300 nM with an injection rate
milk in TBS for 1 h at room temperature. Primary anti- of 2.5 μL/min. This concentration of Reelin was previ-
bodies were incubated overnight at 4 C using the follow- ously used in vivo for full length Reelin protein (Rogers
ing antibodies: Dab1 (Millipore, 1:1000), phospho-Dab1 et al., 2013; Weeber et al., 2002). After the injection, the
(Abcam, 1:1000; for detecting Dab1 phosphorylated at needle was left in for 2 min and then removed, and the
residue Y232), ERK1/2 (Cell Signaling, 1:2000), phospho- incision was sutured. Mice were observed post-
ERK1/2 (Cell Signaling, 1:2000; for detecting ERK1 phos- operatively for 2 h in individual cages on a heating pad.
phorylated at residue Thr202 of ERK1 and ERK2 phos- Following injections, animals were allowed to recover for
phorylated at Thr185/Tyr187), Akt (Cell Signaling, 2 days before beginning behavioural testing.
1:1000), and phospho-Akt (Cell Signaling, 1:1000; for
detecting phosphorylation of Akt at Ser473), GSK-3β
(Cell Signaling [3D10 Cat#9832], 1:1000), phospho-GSK- 2.9 | Behavioural experimentation
3β (Cell Signaling [D85E12, Cat# 5558], 1:1000). Actin
(Sigma, 1:3000) was used as a protein control. Mem- For behaviour testing, mice were balanced for gender
branes were washed 3  10 min in TBS with 0.1% Tween and the experimentor was blinded to the treatment/
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MORRILL ET AL. 1661

genotype of the mice. Testing was performed in the fol- no tone, for 3 min, and freezing was assessed. Cued fear
lowing order: Open field, Hidden Platform water maze conditioning was conducted following the contextual
and fear conditioning. trial, wherein the chamber was altered by scent, lighting
and floor texture. The mice were placed into the altered
chamber and given a 3-min habituation phase (no tone)
2.9.1 | Open-field followed by the conditioned stimulus (90-dB tone) for the
last 3 min of the test. Freezing behaviour was recorded
Mice were recorded in an open-field chamber via ANY-Maze software (Stoelting Co.). Freezing was
(40  40  27 cm) for 15 min. Distance travelled, move- assessed as immobility for at least one consecutive
ment and immobility were analysed with the ANY-Maze second.
animal activity system (Stoelting Co.). Comparing time
spent in the perimeter versus time spent in the centre
was calculated to assess anxiety-like behaviour. 2.10 | Statistical analysis

For luciferase activity measurement (Figure 1) and


2.9.2 | Hidden platform water maze western analysis of dose response to R36 (Figure 2),
one-way ANOVA analysis was performed followed by
A platform (9-cm diameter) was submerged just below Tukey’s Multiple Comparison post hoc test (set at a sig-
the surface in a 1.2-m diameter pool filled with water nificance of p ≤ 0.05). A two-way ANOVA followed by
(deep enough that mice could not touch the bottom) that Tukey’s multiple comparison post hoc test (set at a sig-
contained non-toxic white tempera paint to obscure the nificance of p ≤ 0.05) was used to analyse the beha-
platform and allow easy visibility of the mice. Bold basic vioural tasks and western blot data where significance
shaped visual cues were positioned around the room. of genotype and treatment was assessed. The Grubbs
Mice were placed in the pool and allowed to swim for a outlier test was preformed alongside each statistical
maximum of 60 s. During training, if the mice failed to analysis using an alpha = 0.05. All statistical testing
find the platform within 60 s, they were shown to was performed using GraphPad Prism version 9 for
it. Training consisted of four training days with four trials MacOS (GraphPad Software, San Diego California
per day, separated by a 60-min inter-trial interval. On day UDA, www.Graphpad.com).
5, the probe trial was performed. This involved removal
of the platform and recording of the track of each
mouse’s swim pattern for 60 s with the EthoVision XT 3 | RESULTS
video-tracking software (Noldus). Time spent in the
desired platform target quadrant was assessed. For plat- 3.1 | Small Reelin construct, R36,
form crossings, we defined a ‘crossing’ as the animal’s induces ApoER2 clustering
centre-point entering the predefined target
platform zone. We previously described a new luciferase assay to detect
Reelin signalling (Li et al., 2023). Briefly, cells were
cotransfected with one recombinant ApoER2 receptor
2.9.3 | Fear conditioning fused to the N-terminus of luciferase and one ApoER2
receptor fused to the C-terminus of luciferase. We previ-
Was conducted to evaluate fear-based learning and mem- ously showed that full-length Reelin as well as central
ory. The test was conducted in a square sound- fragment R3456 (Figure 1a) significantly increased
attenuation chamber (25 cm  25 cm) with wire-grid ApoER2 dimerization as indicated by increased firefly
flooring. In training, mice were placed in the chamber luminescence activity (Li et al., 2023). In the present
with background white noise and allowed to explore for study, we examined numerous small fragments of Reelin
3 min before the conditioned stimulus was presented (not shown) and found a fragment of interest, R36, con-
(90-dB tone) for 30 s. At 28 s, the unconditioned stimulus sisting of EGF-like domains 3 and 6 (Figure 1a). One-way
(a mild [0.5-mA] foot shock) was administered for a total ANOVA revealed a significant increase in firefly lumines-
of 2 s. After a period of 90 s, a second conditioned stimu- cence activity in presence of R36 to the same extent as
lus/unconditioned stimulus pairing was conducted, fol- R3456 (F3,12 = 157, p = 0.00001) indicating dimerization
lowed by another 90 s period, for a total of 7 min. of ApoER2 (Figure 1b). A basal dimerization of ApoER2
Contextual fear conditioning was conducted 24 h after was also found, consistent with our previous results (Li
the training in the same chamber, with white noise and et al., 2023). Tukey’s HSD test for multiple comparisons
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1662 MORRILL ET AL.

F I G U R E 2 R36 protein induces downstream signalling. Primary cortical neurons (n = 5) at DIV 14 were treated with Reelin fragment
R36 at concentration 250 pM, 500 pM, 1000 pM or none (control) for 1 h, and the cell lysates were immunoblotted. (a) Micrograph of
immunoblot probed with antibody against phosphorylated Dab1 (Y232) and against total Dab1. (b) Quantification of band intensity for
phosphorylation of Dab1 and (c) total Dab1. Results are presented as ratio to actin protein. (d) Analysis of the ratio of pDab1/Dab1.
(e) Micrograph of immunoblot probed with antibody against phosphorylated ERK1/2 and against total ERK1/2. (f) Quantification of band
intensity for phosphorylation of ERK1/2 and (g) total ERK1/2. Results are presented as ratio to actin protein. (h) Analysis of the ratio of
pERK/ERK. (i) Micrograph of immunoblot probed with antibody against phosphorylated AKT and against total AKT. (j) Quantification of
band intensity for phosphorylation of AKT and (k) total AKT. Results are presented as ratio to actin protein. (l) Analysis of the ratio of
pAKT/AKT. (m) Micrograph of immunoblot probed with antibody against phosphorylated GSK-3β and against total GSK-3β.
(n) Quantification of band intensity for phosphorylation of GSK-3β and (o) total GSK-3β. Results are presented as ratio to actin protein.
(p) Analysis of the ratio of pGSK-3β/GSK-3β. Data are shown as mean ± SEM. **p < 0.01, ****p < 0.0001.

found that the mean value of firefly luminescence activity 3.2 | R36 fragment of Reelin evokes
in presence of R36 was not different from R3456 ApoER2-associated intracellular signalling
(p = 0.70, 95% C.I. = 21,062, 45,875). Luminescence
activity observed with R36 and R3456 was significantly We then investigated if R36 could induce downstream
increased when compared with ApoER2-luc only or con- signalling in vitro. Primary cortical neurons were treated
trol (R36 vs. control, p = 0.0001, C.I. = 235,073, with R36 at concentration 250 pM, 500 pM, 1000 pM or
168,137; R36 vs. ApoER2luc only p = 0.0001, C.I. none (control) for 1-h and the cell lysates were immuno-
= 181,653, 114,717; R3456 vs. control, p = 0.0001, blotted for total and phosphorylated forms of Dab1, AKT,
C.I. = 222,667, 155,731; R3456 vs. ApoER2luc only ERK and GSK-3β (Figure 2a,e,i,m). These proteins are
p = 0.0001, C.I. = 169,247, 102,310). These results known to be involved in Reelin downstream signalling.
indicate that R36, like R3456, is able to induce dimeriza- One-way ANOVA revealed an effect of treatment for
tion of Reelin receptor ApoER2. phosphorylated Dab1 (Figure 2b, F3,14 = 9.013,
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MORRILL ET AL. 1663

p = 0.0014) and ratio of pDab1/Dab1 (Figure 2d, 3.3 | R36 protein injection behavioural
F3,15 = 4.685, p = 0.02). We found no significant effect analysis
on total Dab1 (Figure 2c, F3,16 = 1.003, p = 0.41).
Tukey’s HSD test for multiple comparisons showed a Next, we sought to determine if R36 had a biological
significant increase in phosphorylated Dab1 at high effect in vivo. We used heterozygous reeler mice (HRM)
levels of R36 (1000 pM dose) when compared with con- that exhibit a 50% reduction in Reelin levels together
trol ( p = 0.005, 95% C.I. = 0.50, 0.08), 250-pM dose with cognitive deficits. As depicted in Figure 3a, HRM
( p = 0.003, 95% C.I. = 0.61, 0.12) or 500 pM dose and wildtype control mice aged 12–14 weeks old (n = 10/
( p = 0.007, 95% C.I. = 0.49, 0.07) with no significant group) were injected intracerebroventricularly with
impact on total Dab1 or ratio of pDab1/Dab1 when the either R36 (2-μL at 300-nM) or PBS (control). Forty eight
different concentration of R36 were compared with con- hours following surgery, mice underwent behavioural
trol (Figure 2a–d). testing. First, mice were tested in the open field for gen-
Similarly, one-way ANOVA revealed an effect of eral locomotor activity and anxiety like behaviour. Two-
treatment for phosphorylated ERK1/2 (Figure 2f, way ANOVA showed a significant effect of genotype
F3,16 = 24.64, p = 0.0001) and ratio of pERK/ERK (F1,36 = 7.7, p = 0.009) and treatment (F1,36 = 6.6,
(Figure 2h, F3,16 = 18.33, p = 0.0001). We found no sig- p = 0.01) with no interaction (F1,36 = 3.5, p = 0.07) for
nificant effect on total ERK (Figure 2g, F3,16 = 0.65, total distance travelled (Figure 3b). Tukey’s HSD test for
p = 0.59). Tukey’s HSD test for multiple comparisons multiple comparisons showed a baseline locomotion
showed a significant increase in phosphorylated abnormality in HRM PBS injected group with reduced
ERK1/2 at high levels of R36 (1000 pM dose) when com- total distance travelled compared with WT mice (HRM
pared with control ( p = 0.0001, 95% C.I. = 0.51, PBS vs. WT PBS p = 0.01, 95% C.I. = 8.11, 0.80; HRM
0.26) but not for the 250-pM dose ( p = 0.24) or PBS vs. WT R36 p = 0.02, 95% C.I. = 7.9, 0.60,
500-pM dose ( p = 0.45). We found no significant Figure 3b). There was no significant difference in total
changes in the levels of total ERK1/2. The ratio of time travelled between HRM mice that received the R36
pERK/ERK was significantly increased at high levels of protein injection and the WT animals (HRM R36 vs. WT
R36 (1000-pM dose) when compared with control PBS p = 0.99, 95% C.I. = 3.85, 3.46; HRM R36 vs. WT
( p = 0.0001, 95% C.I. = 0.61, 0.27) but not for the R36 p = 0.91, 95% C.I. = 4.52, 2.79) indicating a rescue
250-pM dose ( p = 0.42) or 500-pM dose ( p = 0.54, of this phenotype.
Figure 2e–h). No differences were observed between the WT mice
One-way ANOVA revealed an effect of treatment for injected with R36 protein and the WT control group
phosphorylated AKT (Figure 2j, F3,16 = 3.88, p = 0.03), (p = 0.96, 95% C.I. = 4.32, 2.98, Figure 3b). Two-way
total AKT (Figure 2k, F3,16 = 4.5, p = 0.02) and ratio of ANOVA revealed no significant difference on time in the
pAKT/AKT (Figure 2l, F3,16 = 9.37, p = 0.0008). Tukey’s centre between groups (Figure 3c; genotype F1,36 = 0.25,
HSD test for multiple comparisons failed to show a signifi- p = 0.62, treatment F1,36 = 0.49, p = 0.49).
cant difference when comparing the different doses to con- Next, we examined spatial learning and memory with
trol for phosphorylated AKT. We found a significant the hidden platform water maze task. For all groups, the
decrease in total AKT (p = 0.008, 95% C.I. = 0.06, 0.39) average slope of the training graph (over the four training
and increase in the ratio of pAKT/AKT (p = 0.007, 95% days) was similar between all groups, suggesting that at
C.I. = 0.88, 0.13) when comparing high dose of R36 the end of training all groups had established a search
against control. strategy to locate the platform (data not shown). During
Finally, one-way ANOVA revealed an effect of treat- the probe trial 24 h after training (that test for spatial
ment for phosphorylated GSK-3β (Figure 2n, F3,16 = 5.84, memory), two-way ANOVA revealed a significant effect
p = 0.009), and ratio of pGSK-3β/GSK-3β (Figure 2o, of genotype (F1,36 = 19.43, p = 0.0001) and treatment
F3,16 = 8.7, p = 0.001) with no effect on total GSK-3β (F1,36 = 10.34, p = 0.003) as well as interaction
(Figure 2p, F3,16 = 0.94, p = 0.44). Tukey’s HSD test for (F1,36 = 6.2, p = 0.02) for time spent in quadrant
multiple comparisons failed to show a significant differ- (Figure 3d). Tukey’s HSD test for multiple comparisons
ence when comparing the different doses to control for revealed that HRM-PBS group spent significantly less
phosphorylated or total GSK-3β. We found a significant time in the target quadrant compared with the WT-PBS
increase in the ratio of pGSK-3β/GSK-3β (p = 0.02, 95% group, demonstrating a baseline deficit in spatial learning
C.I. = 0.68, 0.064) when comparing high dose of R36 and memory ( p = 0.0001, 95% C.I. = 26.35, 7.61). In
against control. contrast, the HRM-R36 group spent significantly more
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1664 MORRILL ET AL.

F I G U R E 3 R36 protein rescues locomotor and learning deficits in HRM. (a) Experimental design. (b) Total distance travelled and
(c) time in the centre, during open field test. (d) Time spent in quadrant (percentage of total time), (e) number of target platform crossing
and (f, g) micrograph representation of averaged heat maps, during Morris water maze. Percent time freezing during (h) contextual and
(i) cued fear conditioning in mice aged 12–14 weeks: HRM given the PBS injection (5M, 5F), HRM given the R36 protein injection (5M, 5F),
WT mice given the PBS injection (6M, 4F) and WT mice given the R36 protein injection (6M, 4F). Data presented as mean ± SEM. For all
groups, n = 10, *p < 0.05, **p < 0.01, ****p < 0.0001.
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MORRILL ET AL. 1665

time in the target quadrant compared with the HRM-PBS contextual test, suggesting that the R36 injection
group ( p = 0.0001, 95% C.I. = 26.35, 7.61) decreased the amount of freezing during contextual fear
(Figure 3d). The HRM-R36 group behaved similar to both conditioning in WT animals. Upon examining the cued-
WT groups (there was no effect of treatment in the WT based test, no baseline deficit was observed in the HRM
group [PBS vs. R36, p = 0.96, 95% C.I. = 11.13, 7.60]). given the PBS injection and overall, no significant differ-
Similarly, two-way ANOVA revealed a significant effect ences were observed amongst groups (two-way ANOVA
of genotype (F1,36 = 4.17, p = 0.04) and treatment showed no significance for genotype: F1,36 = 1.13,
(F1,36 = 5.73, p = 0.02) as well as interaction p = 0.29; or treatment F1,36 = 2.5, p = 0.12, Figure 3i).
(F1,36 = 5.73, p = 0.02) for target platform crossings
(Figure 3e). Tukey’s HSD analysis showed that PBS
injected HRM demonstrated reduced target platform 3.4 | Western blot analysis
crossings compared with the WT animals (vs. WT PBS
p = 0.02, 95% C.I. = 11.71, 0.89; vs. WT R36 p = 0.02, Following the completion of our behavioural assays, tis-
95% C.I. = 11.71, 0.89). Treatment with R36 rescued sue was collected, hippocampal brain tissue was homoge-
this phenotype in HRM (vs. WT PBS p = 0.99, 95% C.I. nized and probed for PSD95 and synaptophysin levels.
= 4.91, 5.91; vs. WT R36 p = 0.99, 95% C.I. = 4.91, No differences were observed between all groups for
5.91; vs. HRM PBS p = 0.009, 95% C.I. = 12.21, 1.39). synaptophysin levels (two-way ANOVA no genotype
Averaged heat maps are shown for each group which F1,12 = 0.38, p = 0.55 or treatment F1,12 = 0.69, p = 0.42
corroborate visually what is observed with total time effect, Figure 4a,b). However, we found a genotype effect
spent in the target quadrant and total platform crossings (two-way ANOVA F1,12 = 18.34, p = 0.001) and treat-
(Figure 3f,g). The difference in spatial memory between ment effect (F1,12 = 8.6, p = 0.01) for PSD95 levels
the HRM group treated with PBS and the other groups is (Figure 4c,d). There was an observed baseline deficit in
easily visualized, with the HRM-PBS group showing no PSD95 levels for the HRM-PBS group when compared
clear quadrant preference and a search pattern encom- with WT groups (vs. WT PBS p = 0.0005, 95% C.I.
passing the majority of the pool with a preference for the = 0.89, 0.28; vs. WT R36 p = 0.001, 95% C.I. = 0.84,
edges of the pool (Figure 3g). In contrast, the HRM group 0.22, Figure 4c,d). HRM that received the R36 protein
given the R36 injection demonstrates a heat map similar had similar levels of PSD95 when compared with WT
to those seen for the WT groups (Figure 3f,g). These data groups that received either the R36 protein ( p = 0.97,
suggest rescue of cognitive deficits in the HRM with the 95% C.I. = 0.35, 0.26) or PBS injection (p = 0.97, 95%
R36 protein. C.I. = 0.35, 0.26) and significantly different from HRM
Hippocampal-dependent associative learning was PBS group ( p = 0.002, 95% C.I. = 0.79, 0.18), suggest-
assessed using a standard contextual fear conditioning ing a rescue in PSD95 levels.
paradigm. In the contextual test, we found an effect of
genotype (F1,36 = 10.89, p = 0.002), treatment
(F1,36 = 5.92, p = 0.02) with interaction (F1,36 = 63.91, 4 | DISCUSSION
p = 0.0001) using two-way ANOVA analysis (Figure 3h).
HRM-PBS mice showed a baseline reduction in the per- It has been well established that Reelin binds to two lipo-
centage of total time spent freezing when compared with protein receptors, VLDLR and ApoER2, causing the
both WT groups (Tukey’s HSD test for multiple compari- receptors to cluster into dimers or higher-order multi-
son; HRM PBS vs. WT PBS p = 0.0001, 95% C.I. mers (D’Arcangelo et al., 1999; Divekar et al., 2014;
= 30.92, 14.38; HRM PBS vs. WT R36 p = 0.001, 95% Dlugosz et al., 2019; Hiesberger et al., 1999; Strasser
C.I. = 20.73, 4.18) (Figure 3h). The HRM-R36 group et al., 2004). Activation of Dab1 induced by ApoER2 clus-
demonstrated a significant recovery of the time spent tering upon Reelin binding is critical for Reelin intracel-
freezing in the contextual test when compared with the lular signalling (Bock & Herz, 2003; Bock & May, 2016;
HRM group given the PBS injection (p = 0.0001, 95% Strasser et al., 2004) together with phosphorylation of
C.I. = 30.92, 14.38, Figure 3h). Following R36 protein ERK1/2 and AKT (Lee et al., 2014; Simo et al., 2007).
injection, the HRM group performed similarly to that of Our laboratory has previously published on a novel
the WT groups given the PBS injection (p = 0.92, 95% split-luciferase assay of ApoER2 clusterin (Li et al., 2023).
C.I. = 10.16, 10.39) suggesting that the R36 protein Using this assay method, we previously investigated the
injection rescued the baseline reduction in time spent Reelin fragment R3456, known to be critical for Reelin
freezing during the contextual test (Figure 3h). We found intracellular signalling. We demonstrated that incubation
a difference between the WT-R36 group and the WT-PBS with R3456 elicited a similar response to full length
group (p = 0.001, 95% C.I. = 19.53, 3.87) in the Reelin in the luciferase assay, indicating receptor
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1666 MORRILL ET AL.

F I G U R E 4 R36 protein injection


enhances markers of post-synaptic
density in the HRM. (a) Anti-
synaptophysin western blot of
hippocampal samples from WT and
HRM groups that received PBS or R36
protein injections. (b) Quantification of
western blot probed for anti-
synaptophysin. Results are presented as
ratio to revert total protein. (c) Anti-
PSD95 western blot of hippocampal
samples from WT or HRM groups that
received PBS or R36 protein injections.
(d) Quantification of western blot
probed for anti-PSD95. Results are
presented as ratio to revert total protein.
n = 4, **p < 0.01. Data shown as mean
± SEM.

dimerization (Li et al., 2023). We further identified down- intracellular signalling pathway, indicated by elevated
stream activation of the Reelin signalling pathway. Con- Dab1 and ERK1/2 phosphorylation in primary cortical
sistent with work from others, we reported that this neurons (Figure 2a–h). Moreover, we observed an
central fragment of Reelin, R3456, can induce Dab1 phos- increase in phosphorylated AKT to total AKT
phorylation (pDab1) in primary neurons (Jossin (Figure 2i–l) and a decrease in active GSK-3β via phos-
et al., 2004; Lee et al., 2014; Li et al., 2023; Nogi phorylation (Figure 2m–p). In neurons, GSK-3β regulates
et al., 2006). Our recent work reveals that R3456 can also various processes critical in functional synaptic plasticity,
activate ERK and AKT (Li et al., 2023). More impor- highlighting GSK-3β as a key signalling molecule in nor-
tantly, we showed rescue of cognitive deficits in HRM mal brain functioning (Jaworski et al., 2019). Our results
with the injection of R3456 protein. suggest that R36 is consistent with full-length Reelin and
Given our success in supplementing R3456 into the R3456 in activating this signalling pathway (Li
HRM (Li et al., 2023), we wanted to further explore the et al., 2023). To further explore the functional efficacy of
possibility of Reelin as a therapeutic to aid in disease the R36 fragment, we performed a single ICV injection of
states affected by reduced Reelin expression and signal- the R36 protein in the HRM and WT mice. As previously
ling such as schizophrenia, autism, depression, epilepsy shown in our study with R3456 supplementation (Li
and Alzheimer’s disease (Alcantara et al., 1998; et al., 2023), HRM control exhibited a deficiency in loco-
D’Arcangelo et al., 1995, 1997; Jossin, 2020; Jossin & motor activity during open field, indicated by a decrease
Goffinet, 2007; Nakajima et al., 1997). Full-length Reelin in total distance travelled when comparing HRM PBS to
(450 kDa) and R3456 (190 kDa) are large protein mol- WT animals (Figure 3b). We did not evidence any differ-
ecules which pose issues for production and cost when ences between groups on anxiety-like behaviour, as all
considering their use as therapeutics. We hypothesized the treatment groups spend the same amount of time in
that if there was a smaller fragment of Reelin, capable of the centre (Figure 3c). In addition, heterozygous reeler
eliciting a similar effect as R3456 and full-length Reelin, mice displayed a deficit in spatial learning as shown as
it could potentially offer a viable gene therapy approach. decreased time spent in quadrant (Figure 3d) and
Using our luciferase assay, we screened numerous Reelin decreased target platform crossings (Figure 3e) during
fragments and found that the construct consisting of Morris water maze. Finally, HRM exhibited a decrease in
EGF-like domains 3 and 6 (R36) (Figure 1a) induced associative learning, evidenced by a decrease in time
ApoER2 dimerization or clustering (Figure 1b). Impor- freezing when compared with WT animals during con-
tantly, R36 also demonstrated activation of the Reelin textual but not cued fear conditioning (Figure 3h,i). Our
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MORRILL ET AL. 1667

results on spatial learning during Morris water maze homozygous reeler mice. In hippocampal cultures, this
were consistent with another model of Reelin deficiency reduction was restored with treatment of full-length
(Paylor et al., 1999). Nonetheless, our data on locomotor Reelin (Niu et al., 2008). To better understand the results
activity and spatial learning deficiencies in the HRM are we observed with treatment of R36 protein, we chose to
not in accordance with studies from other teams, which examine PSD-95 and synaptophysin, markers of post-
did not find any difference between wildtypes animals and pre-synaptic proteins, respectively. We demon-
and HRM in total distance travelled during open field or strated that HRM contain a baseline reduction in levels
learning during Morris water maze (Qiu et al., 2006; of post-synaptic protein-PSD95 and following supple-
Teixeira et al., 2011). This difference might be attributed mentation of R36 protein, we were able to recover the
to the age of the animals, which were much younger than levels of PSD95 in HRM (Figure 4c,d). For synaptophy-
in the experiment described here (6 weeks in Qiu sin, we did not observe a baseline reduction or any
et al., 2006, and 8 weeks in Teixeira et al., 2011, vs. 12– change in levels of protein between any of the groups
14 weeks old in the current experiment). However, there (Figure 4a,b). The results we observed in pre- and post-
seem to be a consensus on associative learning deficiency. synaptic markers with Reelin supplementation may
Deficit in contextual fear conditioning, but not cued, in indicate an improvement in dendritic spine maturation,
HRM control mice when compared with wild type was but further work needs to be done to better visualize
consistent with other reports, even in the younger mice and quantify any effects on dendritic spine density and
(Qiu et al., 2006; Rogers et al., 2013; Weeber et al., 2002). maturation. It has been shown previously that exoge-
We demonstrate that a single administration of the R36 nous Reelin supplementation may increase the number
protein in the HRM was able to recover behavioural defi- of puncta and overall dendritic spine density, future
cits in the open field maze (Figure 3b), the hidden plat- work could include Golgi staining to better visualize
form water maze (Figure 3d,e) and associative fear spine dynamics and localization of pre- and post-
conditioning (Figure 3h). This supports the use of R36, synaptic markers.
essentially as an analogue of Reelin, for therapeutically In summary, we show that a novel construct encom-
increasing Reelin signalling in vivo. Our results are con- passing the Reelin EGF-like repeats 3 and 6 can elicit
sistent with previous work on full length Reelin single receptor dimerization and downstream signalling. We
injection and R3456 single injection in this mouse model demonstrate that a single injection of this R36 protein
(Li et al., 2023; Qiu et al., 2006; Weeber et al., 2002). directly into the brains of HRM rescues spatial learning
Support for Reelin therapies is growing as more and memory deficits and associative learning deficits. We
groups investigate the effects of Reelin supplementa- believe that this supports the potential use of R36 as a
tion and enhancement as recently outlined in a review therapeutic approach for the treatment of diseases which
by Tsuneura et al. (2022). Treatment with Reelin has exhibit reduced Reelin signalling. Additionally, we
been demonstrated to increase dendritic spine density acknowledge that when considering the practicality of
in primary cultured hippocampal neurons (Kim Reelin supplementation as a therapeutic, the use of a pro-
et al., 2015). Others have reported benefits of Reelin tein therapy such as R36 injection, is likely impractical as
supplementation in a mouse model of Angelman syn- this would involve repeated cerebral spinal fluid (CSF)
drome and in WT mice (Rogers et al., 2011, 2013) as injections. Increasingly, gene therapy approaches using
well as in a model of maternal induced activation of adeno-associated virus (AAV)-based vectors are showing
schizophrenia (Ibi et al., 2020). Additionally, our labo- promise in offering therapeutic treatment to a broad
ratory has reported that administration of R3456 into array of diseases (Orlowski et al., 2020). A gene therapy
HRM and a mouse model of fragile X syndrome approach would offer a longer-term application. Future
induced recovery of known cognitive deficits (Li work should consider exploring AAV constructs to better
et al., 2023; Morrill et al., 2022). understand the plausibility and effectiveness of R36 as a
In the literature, exogenous supplementation with potential long-term therapeutic approach.
Reelin has been linked to increased dendritic spine den-
sity in primary hippocampal neurons and increased AUTHOR CONTRIBUTIONS
puncta numbers of synaptophysin and PSD95. It has been Kevin R. Nash and Edwin J. Weeber designed research;
proposed that the Ca2+/calmodulin-dependent protein Nicole K. Morrill and Qingyou Li conducted research;
kinase II β subunit may be necessary for these effects of Kevin R. Nash, Nicole K. Morrill and Aurelie Joly-Amado
exogenous Reelin supplementation on dendritic spine analysed data, performed statistical analysis and wrote
density (Kim et al., 2015). Previous groups have demon- the paper. Kevin R. Nash and Edwin J. Weeber had pri-
strated a baseline reduction in dendritic spine density in mary responsibility for final content. All authors read
hippocampal pyramidal neurons of heterozygous and and approved the final manuscript.
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1668 MORRILL ET AL.

ACK NO WLE DGE MEN TS lamination. The Journal of Biological Chemistry, 278, 38772–
This research did not receive any specific grant from 38779. https://doi.org/10.1074/jbc.M306416200
funding agencies in the public, commercial or not-for- Bock, H. H., & May, P. (2016). Canonical and non-canonical Reelin
signaling. Frontiers in Cellular Neuroscience, 10, 166. https://
profit sectors.
doi.org/10.3389/fncel.2016.00166
Boyle, M. P., Bernard, A., Thompson, C. L., Ng, L., Boe, A.,
CONFLICT OF INTEREST STATEMENT Mortrud, M., Hawrylycz, M. J., Jones, A. R., Hevner, R. F., &
The authors declare no conflicts of interest. Lein, E. S. (2011). Cell-type-specific consequences of Reelin
deficiency in the mouse neocortex, hippocampus, and amyg-
DATA AVAILABILITY STATEMENT dala. The Journal of Comparative Neurology, 519, 2061–2089.
Data described in the manuscript, code book and analytic https://doi.org/10.1002/cne.22655
Cuchillo-Ibanez, I., Balmaceda, V., Mata-Balaguer, T., Lopez-
code will be made available upon request pending
Font, I., & Saez-Valero, J. (2016). Reelin in Alzheimer’s dis-
(e.g., application and approval, payment and other).
ease, increased levels but impaired signaling: When more is
less. Journal of Alzheimer’s Disease, 52, 403–416. https://doi.
P EE R R EV IE W org/10.3233/JAD-151193
The peer review history for this article is available at Curran, T., & D’Arcangelo, G. (1998). Role of reelin in the control
https://www.webofscience.com/api/gateway/wos/peer- of brain development. Brain Research. Brain Research Reviews,
review/10.1111/ejn.15971. 26, 285–294. https://doi.org/10.1016/S0165-0173(97)00035-0
D’Arcangelo, G., Homayouni, R., Keshvara, L., Rice, D. S.,
Sheldon, M., & Curran, T. (1999). Reelin is a ligand for lipo-
R EF E RE N C E S protein receptors. Neuron, 24, 471–479. https://doi.org/10.
Alcantara, S., Ruiz, M., D’Arcangelo, G., Ezan, F., de Lecea, L., 1016/S0896-6273(00)80860-0
Curran, T., Sotelo, C., & Soriano, E. (1998). Regional and cellu- D’Arcangelo, G., Miao, G. G., Chen, S. C., Soares, H. D.,
lar patterns of reelin mRNA expression in the forebrain of the Morgan, J. I., & Curran, T. (1995). A protein related to extra-
developing and adult mouse. The Journal of Neuroscience, 18, cellular matrix proteins deleted in the mouse mutant reeler.
7779–7799. https://doi.org/10.1523/JNEUROSCI.18-19-07779. Nature, 374, 719–723. https://doi.org/10.1038/374719a0
1998 D’Arcangelo, G., Nakajima, K., Miyata, T., Ogawa, M.,
Arnaud, L., Ballif, B. A., Forster, E., & Cooper, J. A. (2003). Fyn Mikoshiba, K., & Curran, T. (1997). Reelin is a secreted glyco-
tyrosine kinase is a critical regulator of disabled-1 during brain protein recognized by the CR-50 monoclonal antibody. The
development. Current Biology, 13, 9–17. https://doi.org/10. Journal of Neuroscience, 17, 23–31. https://doi.org/10.1523/
1016/S0960-9822(02)01397-0 JNEUROSCI.17-01-00023.1997
Ballif, B. A., Arnaud, L., & Cooper, J. A. (2003). Tyrosine phosphor- Divekar, S. D., Burrell, T. C., Lee, J. E., Weeber, E. J., &
ylation of Disabled-1 is essential for Reelin-stimulated activa- Rebeck, G. W. (2014). Ligand-induced homotypic and hetero-
tion of Akt and Src family kinases. Brain Research. Molecular typic clustering of apolipoprotein E receptor 2. The Journal of
Brain Research, 117, 152–159. https://doi.org/10.1016/S0169- Biological Chemistry, 289, 15894–15903. https://doi.org/10.
328X(03)00295-X 1074/jbc.M113.537548
Barr, A. M., Fish, K. N., Markou, A., & Honer, W. G. (2008). Het- Dlugosz, P., Tresky, R., & Nimpf, J. (2019). Differential action of
erozygous reeler mice exhibit alterations in sensorimotor gat- Reelin on oligomerization of ApoER2 and VLDL receptor in
ing but not presynaptic proteins. The European Journal of HEK293 cells assessed by time-resolved anisotropy and fluo-
Neuroscience, 27, 2568–2574. https://doi.org/10.1111/j.1460- rescence lifetime imaging microscopy. Frontiers in Molecular
9568.2008.06233.x Neuroscience, 12, 53. https://doi.org/10.3389/fnmol.2019.00053
Beffert, U., Nematollah Farsian, F., Masiulis, I., Hammer, R. E., Falconer, D. S. (1951). Two new mutants, ‘trembler’ and ‘reeler’,
Yoon, S. O., Giehl, K. M., & Herz, J. (2006). ApoE receptor with neurological actions in the house mouse (Mus musculus
2 controls neuronal survival in the adult brain. Current Biol- L.). Journal of Genetics, 50, 192–201. https://doi.org/10.1007/
ogy, 16, 2446–2452. https://doi.org/10.1016/j.cub.2006.10.029 BF02996215
Beffert, U., Weeber, E. J., Morfini, G., Ko, J., Brady, S. T., Fatemi, S. H., Snow, A. V., Stary, J. M., Araghi-Niknam, M.,
Tsai, L. H., Sweatt, J. D., & Herz, J. (2004). Reelin and cyclin- Reutiman, T. J., Lee, S., Brooks, A. I., & Pearce, D. A. (2005).
dependent kinase 5-dependent signals cooperate in regulating Reelin signaling is impaired in autism. Biological Psychiatry,
neuronal migration and synaptic transmission. The Journal of 57, 777–787. https://doi.org/10.1016/j.biopsych.2004.12.018
Neuroscience, 24, 1897–1906. https://doi.org/10.1523/ Folsom, T. D., & Fatemi, S. H. (2013). The involvement of Reelin in
JNEUROSCI.4084-03.2004 neurodevelopmental disorders. Neuropharmacology, 68, 122–
Bock, H. H., & Herz, J. (2003). Reelin activates SRC family tyrosine 135. https://doi.org/10.1016/j.neuropharm.2012.08.015
kinases in neurons. Current Biology, 13, 18–26. https://doi.org/ Green-Johnson, J. M., Zalcman, S., Vriend, C. Y., Nance, D. M., &
10.1016/S0960-9822(02)01403-3 Greenberg, A. H. (1995). Suppressed T cell and macrophage
Bock, H. H., Jossin, Y., Liu, P., Forster, E., May, P., function in the “reeler” (rl/rl) mutant, a murine strain with
Goffinet, A. M., & Herz, J. (2003). Phosphatidylinositol elevated cerebellar norepinephrine concentration. Brain,
3-kinase interacts with the adaptor protein Dab1 in response Behavior, and Immunity, 9, 47–60. https://doi.org/10.1006/
to Reelin signaling and is required for normal cortical brbi.1995.1005
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MORRILL ET AL. 1669

Hamburgh, M. (1963). Analysis of the postnatal developmental development. The Journal of Neuroscience, 23, 9953–9959.
effects of “Reeler,” a neurological mutation in mice. A study in https://doi.org/10.1523/JNEUROSCI.23-30-09953.2003
developmental genetics. Developmental Biology, 8, 165–185. Kim, M., Jeong, Y., & Chang, Y. C. (2015). Extracellular matrix pro-
https://doi.org/10.1016/0012-1606(63)90040-X tein reelin regulate dendritic spine density through CaMKII-
Hiesberger, T., Trommsdorff, M., Howell, B. W., Goffinet, A., beta. Neuroscience Letters, 599, 97–101. https://doi.org/10.
Mumby, M. C., Cooper, J. A., & Herz, J. (1999). Direct binding 1016/j.neulet.2015.05.033
of Reelin to VLDL receptor and ApoE receptor 2 induces tyro- Lambert de Rouvroit, C., de Bergeyck, V., Cortvrindt, C., Bar, I.,
sine phosphorylation of disabled-1 and modulates tau phos- Eeckhout, Y., & Goffinet, A. M. (1999). Reelin, the extracellu-
phorylation. Neuron, 24, 481–489. https://doi.org/10.1016/ lar matrix protein deficient in reeler mutant mice, is processed
S0896-6273(00)80861-2 by a metalloproteinase. Experimental Neurology, 156, 214–217.
Hong, S. E., Shugart, Y. Y., Huang, D. T., Shahwan, S. A., https://doi.org/10.1006/exnr.1998.7007
Grant, P. E., Hourihane, J. O., Martin, N. D., & Walsh, C. A. Lee, G. H., Chhangawala, Z., von Daake, S., Savas, J. N., Yates, J. R.
(2000). Autosomal recessive lissencephaly with cerebellar 3rd, Comoletti, D., & D’Arcangelo, G. (2014). Reelin induces
hypoplasia is associated with human RELN mutations. Nature Erk1/2 signaling in cortical neurons through a non-canonical
Genetics, 26, 93–96. https://doi.org/10.1038/79246 pathway. The Journal of Biological Chemistry, 289, 20307–
Howell, B. W., Herrick, T. M., & Cooper, J. A. (1999). Reelin- 20317. https://doi.org/10.1074/jbc.M114.576249
induced tyrosine [corrected] phosphorylation of disabled 1 dur- Lee, G. H., & D’Arcangelo, G. (2016). New insights into Reelin-
ing neuronal positioning. Genes & Development, 13, 643–648. mediated signaling pathways. Frontiers in Cellular Neurosci-
https://doi.org/10.1101/gad.13.6.643 ence, 10, 122. https://doi.org/10.3389/fncel.2016.00122
Howell, B. W., Herrick, T. M., Hildebrand, J. D., Zhang, Y., & Li, Q., Morrill, N., Moerman-Herzog, A. M., Barger, S. W., Joly-
Cooper, J. A. (2000). Dab1 tyrosine phosphorylation sites relay Amado, A., Peters, M., Soueidam, H., Diemler, C.,
positional signals during mouse brain development. Current Prabhudeva, S., Weeber, E. J., & Nash, K. R. (2023). Central
Biology, 10, 877–885. https://doi.org/10.1016/S0960-9822(00) repeat fragment of Reelin leads to active Reelin intracellular
00608-4 signaling and rescues cognitive deficits in a mouse model of
Ibi, D., Nakasai, G., Koide, N., Sawahata, M., Kohno, T., Reelin deficiency. Cellular Signalling. Manuscript Submitted
Takaba, R., Nagai, T., Hattori, M., Nabeshima, T., Meseke, M., Prols, F., Schmahl, C., Seebo, K., Kruse, C., Brandt, N.,
Yamada, K., & Hiramatsu, M. (2020). Reelin supplementation Fester, L., Zhou, L., Bender, R., & Rune, G. M. (2018). Reelin
into the hippocampus rescues abnormal behavior in a mouse and aromatase cooperate in ovarian follicle development. Sci-
model of neurodevelopmental disorders. Frontiers in Cellular entific Reports, 8, 8722. https://doi.org/10.1038/s41598-018-
Neuroscience, 14, 285. https://doi.org/10.3389/fncel.2020.00285 26928-x
Ignatova, N., Sindic, C. J., & Goffinet, A. M. (2004). Characteriza- Morrill, N. K., Joly-Amado, A., Li, Q., Prabhudeva, S.,
tion of the various forms of the Reelin protein in the cerebro- Weeber, E. J., & Nash, K. R. (2022). Reelin central fragment
spinal fluid of normal subjects and in neurological diseases. supplementation improves cognitive deficits in a mouse model
Neurobiology of Disease, 15, 326–330. https://doi.org/10.1016/j. of fragile X syndrome. Experimental Neurology, 357, 114170.
nbd.2003.11.008 https://doi.org/10.1016/j.expneurol.2022.114170
Ishii, K., Kubo, K. I., & Nakajima, K. (2016). Reelin and neuropsy- Nakajima, K., Mikoshiba, K., Miyata, T., Kudo, C., & Ogawa, M.
chiatric disorders. Frontiers in Cellular Neuroscience, 10, 229. (1997). Disruption of hippocampal development in vivo by
https://doi.org/10.3389/fncel.2016.00229 CR-50 mAb against reelin. Proceedings of the National Acad-
Jaworski, T., Banach-Kasper, E., & Gralec, K. (2019). GSK-3beta at emy of Sciences of the United States of America, 94, 8196–8201.
the intersection of neuronal plasticity and neurodegeneration. https://doi.org/10.1073/pnas.94.15.8196
Neural Plasticity, 2019, 4209475. https://doi.org/10.1155/2019/ Niu, S., Yabut, O., & D’Arcangelo, G. (2008). The Reelin signaling
4209475 pathway promotes dendritic spine development in hippocam-
Jossin, Y. (2020). Reelin functions, mechanisms of action and sig- pal neurons. The Journal of Neuroscience, 28, 10339–10348.
naling pathways during brain development and maturation. https://doi.org/10.1523/JNEUROSCI.1917-08.2008
Biomolecules, 10, 964. https://doi.org/10.3390/biom10060964 Nogi, T., Yasui, N., Hattori, M., Iwasaki, K., & Takagi, J. (2006).
Jossin, Y., & Goffinet, A. M. (2007). Reelin signals through phos- Structure of a signaling-competent reelin fragment revealed by
phatidylinositol 3-kinase and Akt to control cortical develop- X-ray crystallography and electron tomography. The EMBO
ment and through mTor to regulate dendritic growth. Journal, 25, 3675–3683. https://doi.org/10.1038/sj.emboj.
Molecular and Cellular Biology, 27, 7113–7124. https://doi.org/ 7601240
10.1128/MCB.00928-07 Ohkubo, N., Lee, Y. D., Morishima, A., Terashima, T., Kikkawa, S.,
Jossin, Y., Ignatova, N., Hiesberger, T., Herz, J., Lambert de Tohyama, M., Sakanaka, M., Tanaka, J., Maeda, N.,
Rouvroit, C., & Goffinet, A. M. (2004). The central fragment of Vitek, M. P., & Mitsuda, N. (2003). Apolipoprotein E and
Reelin, generated by proteolytic processing in vivo, is critical Reelin ligands modulate tau phosphorylation through an apo-
to its function during cortical plate development. The Journal lipoprotein E receptor/disabled-1/glycogen synthase kinase-
of Neuroscience, 24, 514–521. https://doi.org/10.1523/ 3beta cascade. The FASEB Journal, 17, 295–297. https://doi.
JNEUROSCI.3408-03.2004 org/10.1096/fj.02-0434fje
Jossin, Y., Ogawa, M., Metin, C., Tissir, F., & Goffinet, A. M. (2003). Ohshima, T., Ogawa, M., Veeranna, Hirasawa, M.,
Inhibition of SRC family kinases and non-classical protein Longenecker, G., Ishiguro, K., Pant, H. C., Brady, R. O.,
kinases C induce a reeler-like malformation of cortical plate Kulkarni, A. B., & Mikoshiba, K. (2001). Synergistic
14609568, 2023, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/ejn.15971 by Yale University, Wiley Online Library on [29/08/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1670 MORRILL ET AL.

contributions of cyclin-dependant kinase 5/p35 and Reelin/- Strasser, V., Fasching, D., Hauser, C., Mayer, H., Bock, H. H.,
Dab1 to the positioning of cortical neurons in the developing Hiesberger, T., Herz, J., Weeber, E. J., Sweatt, J. D.,
mouse brain. Proceedings of the National Academy of Sciences Pramatarova, A., Howell, B., Schneider, W. J., & Nimpf, J.
of the United States of America, 98, 2764–2769. https://doi.org/ (2004). Receptor clustering is involved in Reelin signaling.
10.1073/pnas.051628498 Molecular and Cellular Biology, 24, 1378–1386. https://doi.org/
Orlowski, A., Katz, M. G., Gubara, S. M., Fargnoli, A. S., 10.1128/MCB.24.3.1378-1386.2004
Fish, K. M., & Weber, T. (2020). Successful transduction with Teixeira, C. M., Martin, E. D., Sahun, I., Masachs, N., Pujadas, L.,
AAV vectors after selective depletion of anti-AAV antibodies Corvelo, A., Bosch, C., Rossi, D., Martinez, A., Maldonado, R.,
by immunoadsorption. Molecular Therapy Methods and Clinin- Dierssen, M., & Soriano, E. (2011). Overexpression of Reelin
cal Development, 16, 192–203. https://doi.org/10.1016/j.omtm. prevents the manifestation of behavioral phenotypes related to
2020.01.004 schizophrenia and bipolar disorder. Neuropsychopharmacol-
Patrylo, P. R., Browning, R. A., & Cranick, S. (2006). Reeler homo- ogy, 36, 2395–2405. https://doi.org/10.1038/npp.2011.153
zygous mice exhibit enhanced susceptibility to epileptiform Tissir, F., & Goffinet, A. M. (2003). Reelin and brain development.
activity. Epilepsia, 47, 257–266. https://doi.org/10.1111/j.1528- Nature Reviews. Neuroscience, 4, 496–505. https://doi.org/10.
1167.2006.00417.x 1038/nrn1113
Paylor, R., Hirotsune, S., Gambello, M. J., Yuva-Paylor, L., Togo, M., Numazawa, A., Shumiya, S., & Osanai, M. (1986). Prolon-
Crawley, J. N., & Wynshaw-Boris, A. (1999). Impaired learning gation of the life of a central nervous system-deficient mouse,
and motor behavior in heterozygous Pafah1b1 (Lis1) mutant the reeler. Jikken Dobutsu, 35, 109–115.
mice. Learning & Memory, 6, 521–537. https://doi.org/10.1101/ Tsuneura, Y., Nakai, T., Mizoguchi, H., & Yamada, K. (2022). New
lm.6.5.521 strategies for the treatment of neuropsychiatric disorders based
Qiu, S., Korwek, K. M., Pratt-Davis, A. R., Peters, M., on Reelin dysfunction. International Journal of Molecular Sci-
Bergman, M. Y., & Weeber, E. J. (2006). Cognitive disruption ences, 23, 1829. https://doi.org/10.3390/ijms23031829
and altered hippocampus synaptic function in Reelin haploin- Weeber, E. J., Beffert, U., Jones, C., Christian, J. M., Forster, E.,
sufficient mice. Neurobiology of Learning and Memory, 85, Sweatt, J. D., & Herz, J. (2002). Reelin and ApoE receptors
228–242. https://doi.org/10.1016/j.nlm.2005.11.001 cooperate to enhance hippocampal synaptic plasticity and
Rogers, J. T., Rusiana, I., Trotter, J., Zhao, L., Donaldson, E., learning. The Journal of Biological Chemistry, 277, 39944–
Pak, D. T., Babus, L. W., Peters, M., Banko, J. L., Chavis, P., 39952. https://doi.org/10.1074/jbc.M205147200
Rebeck, G. W., Hoe, H. S., & Weeber, E. J. (2011). Reelin sup- Yamamoto, T., Setsu, T., Okuyama-Yamamoto, A., & Terashima, T.
plementation enhances cognitive ability, synaptic plasticity, (2009). Histological study in the brain of the
and dendritic spine density. Learning & Memory, 18, 558–564. reelin/Dab1-compound mutant mouse. Anatomical Science Inter-
https://doi.org/10.1101/lm.2153511 national, 84, 200–209. https://doi.org/10.1007/s12565-008-0009-7
Rogers, J. T., Zhao, L., Trotter, J. H., Rusiana, I., Peters, M. M., Yoneshima, H., Nagata, E., Matsumoto, M., Yamada, M.,
Li, Q., Donaldson, E., Banko, J. L., Keenoy, K. E., Nakajima, K., Miyata, T., Ogawa, M., & Mikoshiba, K. (1997).
Rebeck, G. W., Hoe, H. S., D’Arcangelo, G., & Weeber, E. J. A novel neurological mutant mouse, yotari, which exhibits
(2013). Reelin supplementation recovers sensorimotor gating, reeler-like phenotype but expresses CR-50 antigen/reelin. Neu-
synaptic plasticity and associative learning deficits in the het- roscience Research, 29, 217–223. https://doi.org/10.1016/S0168-
erozygous reeler mouse. Journal of Psychopharmacology, 27, 0102(97)00088-6
386–395. https://doi.org/10.1177/0269881112463468
Royaux, I., Lambert de Rouvroit, C., D’Arcangelo, G.,
Demirov, D., & Goffinet, A. M. (1997). Genomic organization
of the mouse reelin gene. Genomics, 46, 240–250. https://doi. How to cite this article: Morrill, N. K., Li, Q.,
org/10.1006/geno.1997.4983 Joly-Amado, A., Weeber, E. J., & Nash, K. R.
Simo, S., Pujadas, L., Segura, M. F., La Torre, A., Del Rio, J. A., (2023). A novel Reelin construct, R36, recovered
Urena, J. M., Comella, J. X., & Soriano, E. (2007). Reelin behavioural deficits in the heterozygous reeler
induces the detachment of postnatal subventricular zone cells
mouse. European Journal of Neuroscience, 57(10),
and the expression of the Egr-1 through Erk1/2 activation.
Cerebral Cortex, 17, 294–303. https://doi.org/10.1093/cercor/
1657–1670. https://doi.org/10.1111/ejn.15971
bhj147

You might also like