Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Dec 19.
Published in final edited form as: Basic Clin Pharmacol Toxicol. 2023 Jul 7;133(4):353–363. doi: 10.1111/bcpt.13917

Generation and initial characterization of mice lacking full-length BAI3 (ADGRB3) expression

Fu Hung Shiu 1,2, Jennifer C Wong 1, Debanjan Bhattacharya 3, Yuki Kuranaga 4, Rashed R Parag 4,5, Haifa A Alsharif 6,7, Sushant Bhatnagar 6,7, Erwin G Van Meir 4,5, Andrew Escayg 1
PMCID: PMC10730119  NIHMSID: NIHMS1950134  PMID: 37337931

Abstract

Brain-specific angiogenesis inhibitor 3 (ADGRB3/BAI3) belongs to the family of adhesion G protein-coupled receptors. It is most highly expressed in the brain where it plays a role in synaptogenesis and synapse maintenance. Genome-wide association studies have implicated ADGRB3 in disorders such as schizophrenia and epilepsy. Somatic mutations in ADGRB3 have also been identified in cancer. To better understand the in vivo physiological role of ADGRB3, we used CRISPR/Cas9 editing to generate a mouse line with a 7-base pair deletion in Adgrb3 exon 10. Western blot analysis confirmed that homozygous mutants (Adgrb3Δ7/Δ7) lack full-length ADGRB3 expression. The mutant mice were viable and reproduced in Mendelian ratios but demonstrated reduced brain and body weights and deficits in social interaction. Measurements of locomotor function, olfaction, anxiety levels and prepulse inhibition were comparable between heterozygous and homozygous mutants and wild-type littermates. Since ADGRB3 is also expressed in organs such as lung and pancreas, this new mouse model will facilitate elucidation of ADGRB3’s role in non-central nervous system-related functions. Finally, since somatic mutations in ADGRB3 were identified in patients with several cancer types, these mice can be used to determine whether loss of ADGRB3 function contributes to tumour development.

Keywords: ADGRB3, AGPCR, BAI3, Schizophrenia, seizure

1 ∣. INTRODUCTION AND BACKGROUND

Brain-specific angiogenesis inhibitor 3 (BAI3/ADGRB3) is a member of the adhesion G protein-coupled receptor (ADGR) family, which is comprised of 33 receptors in humans.1,2 ADGRB3 has a typical 7-transmembrane structure and a large extracellular N-terminus containing four thrombospondin type 1 repeats and a GPCR-autoproteolysis-inducing domain.2-4 In humans, ADGRB3 is mainly expressed in neurons and glial cells of the brain, with lower expression in skeletal muscle, lung, pancreatic β-cells and testes.5 ADGRB3 was initially characterized as a putative anti-angiogenic factor6,7 and subsequently shown to promote myoblast fusion.8,9 The high level of ADGRB3 expression in the brain prompted our characterization of its function in the central nervous system.4 In the mouse brain, Adgrb3 expression peaks at postnatal day 0 (P0), but it is stably expressed throughout adulthood.6,10 Adgrb3 is highly expressed in cerebellar Purkinje cells and excitatory neurons in the hippocampus and cortex of adult mice.10 It is also highly expressed at postsynaptic elements10-12 and has been demonstrated to interact with the reticulon 4 (RTN4) receptor,13 neuronal pentraxins,14 and complement-like proteins C1QL1–4.15,16 In humans, single nucleotide polymorphisms (SNPs) and/or copy number variants (CNVs) in ADGRB3 have been associated with schizophrenia,17 addiction,18 anxiety disorder,19 and epilepsy.20,21 Postmortem brain tissue from male patients with schizophrenia show reduced ADGRB3 mRNA expression levels in the hippocampus.22 Moreover, somatic mutations in ADGRB3 have been identified in patients with several types of cancer including breast, lung and prostate, although whether these are driver or passenger mutations are unknown.23 ADGRB3 expression was also lower in human brain glioma specimens.6

Although SNPs and CNVs within ADGRB3 have been associated with several neurological and psychiatric disorders, the in vivo role of ADGRB3 has only been examined within specific neuronal circuits and brain regions of mice. For example, deletion of Adgrb3 in cerebellar Purkinje cells resulted in a deficit in motor learning as revealed by the horizontal optokinetic response assay, a cerebellum-dependent adaptive learning task.16 In addition, while the deletion of Adgrb3 in the olfactory bulb did not alter olfaction, appetite, or locomotion, it did prevent social olfactory learning.24 To more broadly understand the role of ADGRB3 in the brain, we generated a mouse line with a 7-base pair (bp) deletion in Adgrb3 exon 10 and examined heterozygous (Adgrb3+/Δ7) and homozygous (Adgrb3Δ7/Δ7) mutants for potential alterations in behaviour and seizure susceptibility.

2 ∣. MATERIALS AND METHODS

2.1 ∣. Animals

Recombinant Cas9 protein from Streptococcus pyogenes and a synthetic 19-mer (5′-AUAGUGUCUUGGUGACCU-3′) targeting a region of exon 10 (5′-CCCAGGTCACCAAGA-CACTATT-3′) of the mouse Adgrb3 gene were microinjected into individual fertilized eggs from C57BL/6NCrl mice (Charles River, Strain: 027), and the resulting embryos were implanted into recipient pseudo-pregnant females. Genomic DNA of probable founder pups was sequenced in order to identify mice with mutations in exon 10. A founder pup with a heterozygous 7-bp deletion in exon 10, predicted to result in a frame-shift and stop codon (Figure 1A), was identified and selected for generation of the mutant line. To verify germline transmission, the founder mouse (F0 generation) was crossed with C57BL/6NCrl mice (Charles River, Strain: 027) to generate F1 heterozygous mutants (Adgrb3+/Δ7). Adgrb3+/Δ7 mutants were backcrossed for four generations to reduce the likelihood of off-target substitutions. Adgrb3+/Δ7 mutants were then intercrossed to generate wild-type (WT), heterozygous (Adgrb3+/Δ7) and homozygous (Adgrb3Δ7/Δ7) offspring. Adgrb3Δ7/Δ7 mice were genotyped as previously described.25 Mice were housed in groups of three to five on a 12-h light/dark cycle with standard laboratory rodent chow (Lab Diet, #5001) and water available ad libitum. All experiments were performed by investigators who were blinded to genotype and in accordance with the Emory University Institutional Animal Care and Use Committee (IACUC) guidelines and the Basic & Clinical Pharmacology & Toxicology policy for experimental and clinical studies.26

FIGURE 1.

FIGURE 1

Generation and characterization of Adgrb3Δ7/Δ7 mice. (A) Exon 10 was targeted by CRISPR/Cas9. A 7-bp deletion in exon 10 was confirmed by Sanger sequencing. (B) Western blot shows the absence of full-length ADGRB3 protein (~172 kDa) in Adgrb3Δ7/Δ7 mice. *Smaller ~100, 72, 48 and 27 kDa bands are observed in Adgrb3Δ7/Δ7 mice and WT littermates. Whether these are non-specific or represent shorter isoforms of ADGRB3 remain to be determined. (C) Nissl staining of coronal brain sections show no obvious structural defects in Adgrb3Δ7/Δ7 mice. (D) Matings between Adgrb3+/Δ7 mice yielded offspring of each genotype in the expected Mendelian ratio and a similar ratio of males and females (E).

2.2. ∣. Western blot

Mouse brain tissue was homogenized in lysis buffer (50 mM Tris–HCl [pH 7.4], 150 mM NaCl, 1% [v/v] NP-40, 1 mM EDTA and 10% [v/v] glycerol). Lysates were then centrifuged for 10 min at 13500 × g. The supernatant was collected and protein concentration was determined using a BCA assay (Thermo Scientific, 23225). Laemmli sample buffer (4X) (500 mM Tris–HCl [pH 6.8], 44.4% [v/v] glycerol, 4.4% sodium dodecyl sulfate, 0.02% bromophenol blue) containing β-mercaptoethanol (10 mM) was added into the supernatant. Protein lysates (10 μg) from each mouse were subjected to SDS-PAGE electrophoresis without pre-boiling using an 8% polyacrylamide gel, followed by transfer to PVDF membranes (Bio-Rad, 1620177). Blots were blocked with tris-buffered saline with 0.1% Tween 20 and 5% non-fat dry milk, followed by incubation with a custom-made (GenScript), antigen-purified polyclonal rabbit anti-mouse ADGRB3 C-terminal antibody (epitope: GRREVQDAFRCRLRNCQDPINADSSSSFPNGHAQIMTDFEKDVDIACRSVLHKDIGPCRAATITGTLSRISLNDDEEEKGTNPEGLSYSTLPGNVISKVIIQQPTGLHMPSMNELSNPCLKKENTELRRTVYLCTDDNLRGADMDIVHPQERMMESDYIVMPRSSVSTQPSMKEESKMNIGMETLPHERLLHYKVNPE, 1:3000) or a mouse anti-β-actin antibody (Santa Cruz Biotechnology, sc-69879, 1:3000) overnight. The membrane was subsequently probed with a rabbit HRP tagged secondary antibody (Thermo Scientific, 31460, 1:5000) for ADGRB3 or a mouse HRP tagged secondary antibody (Thermo Scientific, 31430, 1:5000) for β-actin and processed for chemiluminescence detection using ECL (Thermo Scientific, 34580) (n = 3 mice/genotype).

2.3. ∣. Brain and body weight assessment

Male Adgrb3Δ7/Δ7, Adgrb3+/Δ7 and WT littermates (2-3 months old) were weighed and anaesthetised with isoflurane. Brains were harvested and immediately weighed (n = 4–9 mice/genotype).

2.4. ∣. Behaviour assessments

Behavioural experiments were conducted on male Adgrb3Δ7/Δ7, Adgrb3+/Δ7 and WT littermates (3 to 5 months old). Behavioural experiments were performed with two separate cohorts of mice. Cohort 1 was tested for open field and novel object recognition, novel cage and three-chamber social interaction. Cohort 2 underwent prepulse inhibition and the buried food test. All behavioural experiments were video recorded, and videos were analysed with ANY-Maze Video Tracking System (Stoelting Co.).

2.5 ∣. Open field and novel object recognition

Locomotor activity and anxiety levels were assessed by open field. Learning and memory was assessed by novel object recognition. The tasks were performed over a 4-day period as previously described.27,28 The apparatus is comprised of opaque Plexiglas walls (60 cm × 60 cm × 60 cm). On day 1, each mouse was allowed to explore the empty apparatus for 10 min; the total distance travelled and the time spent in the centre were scored as a measure of locomotor activity and anxiety, respectively. On days 2 and 3, two identical objects (cube or sphere) were placed in the centre of the apparatus, and the experimental mouse was allowed 10 min to explore. On day 4, one of the objects from days 2 and 3 was replaced with a novel object (e.g., a sphere was replaced with a cube or vice versa). The objects and the location of the novel versus familiar object were counterbalanced. A discrimination ratio was calculated for each mouse (time exploring the novel object/(time exploring the novel object + time exploring the familiar object) (n = 8–9 mice/genotype).

2.6 ∣. Three-chamber social interaction

Sociability and social discrimination were assessed using the three-chamber social interaction paradigm as previously described.27,28 The experiment consisted of three 10-min trials. In Trial 1, the experimental mouse was placed in the centre chamber, with an empty cylindrical wire cage in the left and right chambers; the mouse was allowed to freely explore. In Trial 2, an age-matched male C57BL/6J mouse (stranger) was placed under one of the wire cages. Sociability was defined as the time spent exploring the stranger mouse versus the empty cage. In Trial 3, a second age- and sex-matched C57BL/6J mouse (novel mouse) was placed under the previously empty wire cage. As a metric of social discrimination, the time spent exploring the familiar versus novel mouse was calculated (n = 8 mice/genotype).

2.7 ∣. Novel cage

Novel cage experiment was performed as previously described.27,28 Each mouse was placed into a novel standard mouse cage (33 cm × 18 cm × 15 cm), and the time spent grooming, digging, rearing and circling were recorded over a 10-min period (n = 7–10 mice/genotype).

2.8 ∣. Prepulse inhibition (PPI)

PPI is a measure of sensorimotor gating, the process by which the brain filters out irrelevant sensory stimulation. It is assessed by measuring the inhibition of the reflex response to a startle stimulus that occurs when a weaker stimulus (prepulse) precedes the startle stimulus. Each mouse was placed in a sound-attenuated chamber (San Diego Instruments). For the startle response, the mouse was exposed to auditory-evoked startle stimuli (120 dB, 20 ms), and its response was measured by a piezoelectric accelerometer. The mouse was then subjected to 12 startle trials (120 dB, 20 ms), 12 prepulse trials (20 ms white noise at 74, 78, 82 or 86 dB at 100-ms intervals) and 12 prepulse/startle trials (20 ms white noise at 74, 78, 82 or 86 dB at 100-ms intervals and 20 ms 120-dB startle stimulus). The startle and prepulse trials were randomized. The percent PPI was calculated [100 — (startle amplitude on prepulse–pulse trials/startle amplitude on pulse alone trials) × 100] for each experimental mouse (n = 11–13 mice/genotype).

2.9 ∣. Buried food test

Olfactory function was examined using the buried food test as described previously.27 Two days before test day, chocolate-flavoured pellets (Bio-Serv F05472-1) were introduced into the home cage of the experimental mice to allow for habituation to the novel and palatable stimulus. Mice were food-deprived 1 day before the test day. On the test day, chocolate pellets were buried in a random corner of a clean mouse cage. The latency for the experimental mouse to find and eat (grasp and bite) the chocolate pellets was recorded. A maximum feeding latency score of 900 s was assigned if the mouse did not feed within 15 min (n = 7–10 mice/genotype).29

2.10 ∣. Seizure induction

Susceptibility to induced seizures was tested in 3- to 5-month-old male and female Adgrb3Δ7/Δ7, Adgrb3+/Δ7 and WT littermates.

2.11 ∣. 6 Hz seizure induction

6 Hz psychomotor seizures were induced as previously described.30-33 A topical analgesic was applied to the cornea (0.5% tetracaine hydrochloride) prior to seizure induction. Experimental mice were subjected to corneal electrical stimulation (3 s at 17 mA for males and 13 mA for females) using a constant current device (ECT Unit 57 800; Ugo Basile, Comerio, Italy). Behavioural seizures were scored on a modified Racine scale: RS0 = no abnormal behaviour; RS1 = immobile ≥ 3 s, RS2 = forelimb clonus, paw waving, RS3 = rearing and falling (n = 8–14 mice/sex/genotype).

2.12 ∣. Flurothyl-induced seizures

Flurothyl seizure induction was performed as previously described.28,33-35 Latencies to the first myoclonic jerk (MJ) and generalized tonic–clonic seizure (GTCS) were recorded (n = 7–9 mice/sex/genotype).

2.13 ∣. Statistical analysis

Prism v 9.3.1 software (GraphPad, San Diego, California) was used for statistical analyses. A two-way ANOVA followed by Sidak’s multiple comparisons test was used to compare sociability, social discrimination and PPI data between each genotype. For novel object recognition, a one-sample parametric t-test was used to compare the time spent with the novel object versus 50% chance. A one-way ANOVA followed by Tukey’s multiple comparisons test was used to analyse brain/body weight measurements, the total distance travelled and total time spent in the centre of the open field, the latency to the MJ and GTCS during flurothyl seizure induction, the startle response and feeding latency. An unpaired nonparametric Mann–Whitney U test was used to compare Racine scores between each genotype after 6 HZ seizure induction. A chi-square test was used to compare the observed number of offspring of each genotype to the expected Mendelian ratio and sex ratio. All results are presented as mean ± SEM, and a p value < 0.05 was considered statistically significant.

3 ∣. RESULTS

3.1 ∣. Generation and characterization of Adgrb3Δ7/Δ7 mice

To generate Adgrb3Δ7/Δ7 mice, we used CRISPR/Cas9 to target exon 10. A founder mouse with a 7-bp deletion (CCCAGGT) in exon 10 (Figure 1A), predicted to result in a frameshift and premature stop codon, was identified and used to generate the mouse line by backcrossing to C57BL/6NCrl for four generations (N4 generation). Male and female Adgrb3+/Δ7 mice at the N4 generation were intercrossed to generate heterozygous and homozygous mutants and WT littermates that were used for all experiments. Western blot analyses showed full-length ADGRB3 protein expression (~172 kDa) was absent in Adgrb3Δ7/Δ7 mutants (Figure 1B). Four additional proteins at ~100, 72, 48 and 27 KDa were also detected but were present in both Adgrb3Δ7/Δ7 mutants and WT littermates. Whether these represent non-specific bands or result from alternative transcripts requires further study. Nissl staining of brain sections did not reveal any gross structural abnormalities between WT littermates and Adgrb3Δ7/Δ7 mutants (Figure 1C). From a total of 31 litters (217 mice), offspring of each genotype was born in the expected 1:2:1 Mendelian ratio (Figure 1D), and an equal ratio of male and female pups was observed (Figure 1E). All genotypes were viable and fertile.

Adgrb3Δ7/Δ7 mutants, followed for over 600 days, displayed no evidence of spontaneous cancer formation, suggesting that gene loss is not sufficient to initiate cancer (data not shown). Aged mice had no obvious visible defects except for the development of severe dermatitis starting at ~250 days in ~40% of the female Adgrb3Δ7/Δ7 mutants, which mandated their termination per IACUC guidelines. No age and sex-matched Adgrb3+/Δ7 mutants or WT littermates developed the same severity of dermatitis as female Adgrb3Δ7/Δ7 mutants.

3.2 ∣. Adgrb3Δ7/Δ7 mice exhibit reduced brain and body weight

We compared brain and body weights of 2- to 3-month-old male mice of each genotype. The average brain weight of Adgrb3+/Δ7 and Adgrb3Δ7/Δ7 mutants was significantly less than WT littermates (Adgrb3+/Δ7, p < 0.05; Adgrb3Δ7/Δ7, p < 0.001), and the average brain weight of Adgrb3Δ7/Δ7 mice was significantly lower than Adgrb3+/Δ7 (p < 0.05) (Figure 2A). The body weight of Adgrb3Δ7/Δ7 mice was also significantly less than WT littermates (p < 0.05) (Figure 2B). However, brain/body weight ratios were similar across genotypes (Figure 2C).

FIGURE 2.

FIGURE 2

Male Adgrb3Δ7/Δ7 mice (2- to 3-month-old) exhibit lower brain and body weight. Adgrb3Δ7/Δ7 and Adgrb3+/Δ7 mice had lower brain (A) and body weight (B) compared to WT littermates. (C) Brain/body weight ratio was comparable across all three genotypes.WT, n = 4; Adgrb3+/Δ7, n = 8; Adgrb3Δ7/Δ7, n = 9. Mean ± SEM. *p < 0.05, ***p < 0.001.

3.3 ∣. Adgrb3Δ7/Δ7 mice exhibit deficits in social discrimination

We observed no statistically significant differences between Adgrb3+/Δ7 and Adgrb3Δ7/Δ7 mutants and WT littermates in measurements of locomotor activity (Figure 3A), anxiety levels (Figure 3B,D), repetitive behaviours (Figure 3E), startle response and percent PPI (Figure 3F,G). All genotypes spent significantly more time exploring the novel object compared to 50% chance in the novel object recognition task (WT, p < 0.001; Adgrb3+/Δ7, p < 0.0001; Adgrb3Δ7/Δ7, p < 0.01), suggesting intact long-term recognition memory (Figure 3C).

FIGURE 3.

FIGURE 3

Adgrb3Δ7/Δ7 mice and WT littermates show comparable performance in open field, novel object recognition, light/dark box, novel cage paradigm and PPI. In the open field task, there was no significant effect of genotype on (A) total distance travelled and (B) total time spent in the centre of the open field. (C) All three genotypes spent significantly more time exploring the novel object, indicating intact object recognition memory. WT, n = 8; Adgrb3+/A7, n = 9; Adgrb3Δ7/Δ7, n = 9. (D) Comparable anxiety levels were observed across all genotypes as assessed by the light/dark box paradigm. (E) In the novel cage paradigm, no significant differences were observed between genotypes in the time spent grooming, digging, rearing and circling. WT, n = 7; Adgrb3+/Δ7, n = 10; Adgrb3Δ7/Δ7, n = 7. Mice of all three genotypes exhibited comparable (F) startle response and (G) prepulse inhibition. WT, n = 13; Adgrb3+/Δ7, n = 11; Adgrb3Δ7/Δ7, n = 11. Mean ± SEM. **p < 0.01, ***p < 0.001, ****p < 0.0001.

In the three-chamber social interaction task, all genotypes spent significantly more time exploring the stranger mouse than the empty cage in the “sociability” trial, demonstrating normal sociability (WT, p < 0.05; Adgrb3+/Δ7, p < 0.01; Adgrb3Δ7/Δ7, p < 0.001) (Figure 4A). During the “social discrimination” trial, WT littermates and Adgrb3+/Δ7 showed a significant preference for the novel mouse compared to the familiar mouse, demonstrating normal social discrimination (WT, p < 0.05; Adgrb3+/A7, p < 0.001). In contrast, Adgrb3Δ7/Δ7 mice did not show a statistically significant preference for the novel mouse, suggesting a deficit in social discrimination (Figure 4B). In the buried food test, the latency to find buried food was comparable across all genotypes (Figure 4C), demonstrating that the observed deficit in social discrimination in the Adgrb3Δ7/Δ7 mutants is unlikely to be due to olfactory dysfunction.

FIGURE 4.

FIGURE 4

Adgrb3Δ7/Δ7mice exhibit deficits in social discrimination. (A) Mice of all genotypes spent more time exploring a stranger mouse than an empty cage (object), demonstrating intact sociability. (B) Adgrb3Δ7/Δ7 mice failed to discriminate between a novel and a familiar mouse, indicating a deficit in social discrimination. WT, n = 8; Adgrb3+/Δ7, n = 8; Adgrb3Δ7/Δ7, n = 8. (C) The latency to find buried food was comparable across genotypes. WT, n = 9; Adgrb3+/Δ7, n = 7; Adgrb3Δ7/Δ7, n = 10. Mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

3.4 ∣. Male Adgrb3Δ7/Δ7 mice show a modest increase in susceptibility to flurothyl-induced seizures

In the 6 Hz paradigm, acute psychomotor seizures were induced via corneal stimulation,36 and seizure severity was scored using a modified Racine scale as we previously described.27 Racine scores were found to be comparable between all genotypes regardless of sex (Figure 5A,D), indicating there were no statistically significant differences in the severity of these behavioural seizures between genotypes. In the flurothyl seizure induction paradigm, acute seizures were induced via inhalation of flurothyl, a GABAa antagonist.37 Male Adgrb3Δ7/Δ7 mutants displayed shorter average latencies to the first myoclonic jerk (MJ) (P < 0.05) (Figure 5B); however, the average latency to the first generalized tonic–clonic seizure (GTCS) was comparable between genotypes (Figure 5C). Regardless of genotype, there were no statistically significant differences in average latencies to the MJ or GTCS among female mice (Figure 5E,F).

FIGURE 5.

FIGURE 5

A modest increase in susceptibility to flurothyl-induced seizures in male Adgrb3Δ7/Δ7 mice. (A and D) Susceptibility to 6 Hz induced seizures was comparable between same-sex mice of all genotypes. Male: WT, n = 10; Adgrb3+/Δ7, n = 8; Adgrb3Δ7/Δ7, n = 10; female: WT, n = 11; Adgrb3+/Δ7, n = 14; Adgrb3Δ7/Δ7, n = 14. (B and E) Male, but not female Adgrb3Δ7/Δ7 mutants exhibited shorter latencies to the first flurothyl-induced myoclonic jerk (MJ). (C and F) Male and female mice of all genotypes exhibited comparable latencies to the first generalized tonic–clonic seizure (GTCS). Male: WT, n = 8; Adgrb3+/Δ7, n = 7; Adgrb3Δ7/Δ7, n = 9; female: WT, n = 8; Adgrb3+/Δ7, n = 8; Adgrb3Δ7/Δ7, n = 8. Mean ± SEM. *p < 0.05.

4 ∣. DISCUSSION

Multiple SNPs and CNVs in ADGRB3 have been implicated in schizophrenia and epilepsy17,20,21,38-40; however, it remains unclear whether ADGRB3 plays a causal role in these disorders. To date, functional analyses of identified SNPs have not been conducted and most CNVs include several adjacent genes. Similarly, somatic mutations were identified in ADGRB3 in several cancers, including lung and ovarian,23 but their significance as disease drivers have not been investigated. A recent study reported a consanguineous family in which two siblings harbored a homozygous intragenic duplication of ADGRB3 introns 3–17. The probands had intellectual disability, cerebellar atrophy, psychosis, epilepsy and anxiety disorder.41 The duplication was predicted to alter the reading frame and reduce ADGRB3 expression via nonsense-mediated decay of the mRNA transcript. However, the authors were unable to directly examine mRNA levels since ADGRB3 is almost exclusively expressed in the brain.41 Thus, to better understand the clinical relevance of reduced ADGRB3 expression, we generated a mouse line that constitutively lacks full-length Adgrb3 gene expression. Western blotting with a C-terminal antibody confirmed that the homozygous mutants lack full-length ADGRB3 protein.

The deletion of Adgrb3 had a modest impact on seizure thresholds, with the only observed statistically significant alteration being a reduction in the latency to the first flurothyl-induced MJ in male Adgrb3Δ7/Δ7 mutants (Figure 5B). In addition to ADGRB3, the ADGRB subfamily also includes ADGRB1 (BAI1) and ADGRB2 (BAI2).3,4 We previously demonstrated that male and female mice lacking full-length Adgrb1 (Adgrb1−/−) exhibit significantly increased susceptibility to flurothyl-and 6 Hz-induced seizures.27 Seizure susceptibility has not yet been examined in Adgrb2−/− mutants.

With the exception of a deficit in social discrimination, we found that the Adgrb3Δ7/Δ7mutants exhibited fairly normal behaviour. We previously reported that Adgrb1−/− mutants show deficits in both social discrimination and sociability.27 While the novel object recognition task revealed deficits in recognition memory in Adgrb1−/− mutants, the performance of Adgrb3Δ7/Δ7 mutants in this task was comparable to their WT littermates. When examined using the Morris water maze test, Adgrb1−/− mutants exhibited deficits in spatial learning and memory,42 while normal performance was observed in Adgrb2−/− mutants.43 Adgrb2−/− mutants also displayed a higher level of hippocampal neurogenesis and greater antidepressant-like behaviour as assessed by the forced swim test.43

The behaviour alterations observed with loss of each member of the ADGRB subfamily might reflect, in part, the impact on different associated G-protein independent signalling pathways. ADGRB1 is a phosphatidylserine mediated phagocytic receptor,44,45 and the loss of ADGRB1 expression leads to reduced phagocytic uptake by glial cells and increased MDM2-mediated p53 degradation, both of which could contribute to the observed abnormalities in social behaviour and memory performance, and increased seizure susceptibility.46-50 ADGRB2 inhibits vascular endothelial growth factor (VEGF),51 which is known to stimulate adult hippocampal neurogenesis and promote antidepressant effects.52 Therefore, the lack of ADGRB2 can lead to the activation of VEGF, which might be the underlying cause of the antidepressant-like behaviour in Adgrb2−/− mice.43

With the exception of the identified deficit in social discrimination, the performance of Adgrb3Δ7/Δ7 mice was similar to WT littermates in the selected behavioural assays. This result was surprising as ADGRB3 is associated with a wide range of psychiatric and neurological disorders and several cancer types.4,6,14,17,20,22,23,41 It is possible that Adgrb3Δ7/Δ7 mutants might exhibit abnormalities in behaviours that were not examined in the current study. For example, mice lacking the ADGRB3 ligand, C1QL3,15 display altered circadian activity,53 and mice lacking C1QL115 exhibit impaired cerebellar motor learning16 and reduced auditory brainstem response during audiometric testing.54 Furthermore, we cannot exclude the possibility that the residual bands detected by Western blotting in Adgrb3Δ7/Δ7 mice might represent functional ADGRB3 protein isoforms. Shorter N-terminal truncated forms of ADGRB1 were recently shown to arise from usage of an alternative promoter in intron 17 of ADGRB1.55 However, these ADGRB1 isoforms did not rescue the neurological deficits found in mice lacking full-length ADGRB1.42 Finally, it should be noted that a mild increase in seizure susceptibility was only observed in male Adgrb3Δ7/Δ7 mice, suggesting possible sex effects. Additional studies using both sexes should be conducted to further explore the role for ADGRB3 in neurological and psychiatric disorders and to examine whether the loss of ADGRB3 might accelerate tumour formation in the context of other genetic alterations.

ACKNOWLEDGEMENTS

We would like to thank Anu Korukonda for assisting with the buried food test.

Funding information

This work was supported by the National Institutes of Health (National Cancer Institute) (EGVM, CA235162) and the Emory Brain Health Center (EGVM, AE). The content is solely the responsibility of the authors and does not necessarily reflect the official views of the National Institutes of Health.

Footnotes

CONFLICT OF INTEREST STATEMENT

All the authors declare no competing interests.

DATA AVAILABILITY STATEMENT

The data that support the findings of this study are available from the corresponding author upon request.

REFERENCES

  • 1.Hamann J, Aust G, Arac D, et al. International Union of Basic and Clinical Pharmacology. XCIV. Adhesion G protein-coupled receptors. Pharmacol Rev. 2015;67(2):338–367. doi: 10.1124/pr.114.009647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lala T, Hall RA. Adhesion G protein-coupled receptors: structure, signaling, physiology, and pathophysiology. Physiol Rev. 2022;102(4):1587–1624. doi: 10.1152/physrev.00027.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cork SM, Van Meir EG. Emerging roles for the BAI1 protein family in the regulation of phagocytosis, synaptogenesis, neurovasculature, and tumor development. J Mol Med (Berl). 2011;89(8):743–752. doi: 10.1007/s00109-011-0759-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Stephenson JR, Purcell RH, Hall RA. The BAI subfamily of adhesion GPCRs: synaptic regulation and beyond. Trends Pharmacol Sci. 2014;35(4):208–215. doi: 10.1016/j.tips.2014.02.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Tan A, Ke S, Chen Y, et al. Expression patterns of C1ql4 and its cell-adhesion GPCR Bai3 in the murine testis and functional roles in steroidogenesis. FASEB j. 2019;33(4):4893–4906. doi: 10.1096/fj.201801620RR [DOI] [PubMed] [Google Scholar]
  • 6.Kee HJ, Ahn KY, Choi KC, et al. Expression of brain-specific angiogenesis inhibitor 3 (BAI3) in normal brain and implications for BAI3 in ischemia-induced brain angiogenesis and malignant glioma. FEBS Lett. 2004;569(1–3):307–316. doi: 10.1016/j.febslet.2004.06.011 [DOI] [PubMed] [Google Scholar]
  • 7.Shiratsuchi T, Nishimori H, Ichise H, Nakamura Y, Tokino T. Cloning and characterization of BAI2 and BAI3, novel genes homologous to brain-specific angiogenesis inhibitor 1 (BAI1). Cytogenet Cell Genet. 1997;79(1–2):103–108. doi: 10.1159/000134693 [DOI] [PubMed] [Google Scholar]
  • 8.Hamoud N, Tran V, Croteau LP, Kania A, Cote JF. G-protein coupled receptor BAI3 promotes myoblast fusion in vertebrates. Proc Natl Acad Sci U S A. 2014;111(10):3745–3750. doi: 10.1073/pnas.1313886111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hamoud N, Tran V, Aimi T, et al. Spatiotemporal regulation of the GPCR activity of BAI3 by C1qL4 and stabilin-2 controls myoblast fusion. Nat Commun. 2018;9(1):4470. doi: 10.1038/s41467-018-06897-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sigoillot SM, Iyer K, Binda F, et al. The secreted protein C1QL1 and its receptor BAI3 control the synaptic connectivity of excitatory inputs converging on cerebellar Purkinje cells. Cell Rep. 2015;10(5):820–832. doi: 10.1016/j.celrep.2015.01.034 [DOI] [PubMed] [Google Scholar]
  • 11.Selimi F, Cristea IM, Heller E, Chait BT, Heintz N. Proteomic studies of a single CNS synapse type: the parallel fiber/Purkinje cell synapse. PLoS Biol. 2009;7(4):e83. doi: 10.1371/journal.pbio.1000083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Collins MO, Husi H, Yu L, et al. Molecular characterization and comparison of the components and multiprotein complexes in the postsynaptic proteome. J Neurochem. 2006;97(Suppl 1):16–23. doi: 10.1111/j.1471-4159.2005.03507.x [DOI] [PubMed] [Google Scholar]
  • 13.Wang J, Miao Y, Wicklein R, et al. RTN4/NoGo-receptor binding to BAI adhesion-GPCRs regulates neuronal development. Cell. 2021;184(24):5869–5885 e25. doi: 10.1016/j.cell.2021.10.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sticco MJ, Pena Palomino PA, Lukacsovich D, et al. C1QL3 promotes cell-cell adhesion by mediating complex formation between ADGRB3/BAI3 and neuronal pentraxins. FASEB j. 2021;35(1):e21194. doi: 10.1096/fj.202000351RR [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bolliger MF, Martinelli DC, Sudhof TC. The cell-adhesion G protein-coupled receptor BAI3 is a high-affinity receptor for C1q-like proteins. Proc Natl Acad Sci U S A. 2011;108(6):2534–2539. doi: 10.1073/pnas.1019577108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kakegawa W, Mitakidis N, Miura E, et al. Anterograde C1ql1 signaling is required in order to determine and maintain a single-winner climbing fiber in the mouse cerebellum. Neuron. 2015;85(2):316–329. doi: 10.1016/j.neuron.2014.12.020 [DOI] [PubMed] [Google Scholar]
  • 17.DeRosse P, Lencz T, Burdick KE, Siris SG, Kane JM, Malhotra AK. The genetics of symptom-based phenotypes: toward a molecular classification of schizophrenia. Schizophr Bull. 2008;34(6):1047–1053. doi: 10.1093/schbul/sbn076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Liu QR, Drgon T, Johnson C, Walther D, Hess J, Uhl GR. Addiction molecular genetics: 639,401 SNP whole genome association identifies many “cell adhesion” genes. Am J Med Genet B Neuropsychiatr Genet. 2006;141B(8):918–925. doi: 10.1002/ajmg.b.30436 [DOI] [PubMed] [Google Scholar]
  • 19.Gonda X, Eszlari N, Torok D, et al. Genetic underpinnings of affective temperaments: a pilot GWAS investigation identifies a new genome-wide significant SNP for anxious temperament in ADGRB3 gene. Transl Psychiatry. 2021;11(1):337. doi: 10.1038/s41398-021-01436-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Albuz B, Ozdemir O, Silan F. The high frequency of chromosomal copy number variations and candidate genes in epilepsy patients. Clin Neurol Neurosurg. 2021;202:106487. doi: 10.1016/j.clineuro.2021.106487 [DOI] [PubMed] [Google Scholar]
  • 21.Leu C, Balestrini S, Maher B, et al. Genome-wide polygenic burden of rare deleterious variants in sudden unexpected death in epilepsy. EBioMedicine. 2015;2(9):1063–1070. doi: 10.1016/j.ebiom.2015.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Unroe KA, Glover ME, Shupe EA, Feng N, Clinton SM. Perinatal SSRI exposure disrupts G protein-coupled receptor BAI3 in developing dentate gyrus and adult emotional behavior: relevance to psychiatric disorders. Neuroscience. Sep 1 2021;471:32–50. doi: 10.1016/j.neuroscience.2021.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kan Z, Jaiswal BS, Stinson J, et al. Diverse somatic mutation patterns and pathway alterations in human cancers. Nature. 2010;466(7308):869–873. doi: 10.1038/nature09208 [DOI] [PubMed] [Google Scholar]
  • 24.Wang CY, Liu Z, Ng YH, Sudhof TC. A synaptic circuit required for acquisition but not recall of social transmission of food preference. Neuron. 2020;107(1):144–157 e4. doi: 10.1016/j.neuron.2020.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bhattacharya D, Van Meir EG. A simple genotyping method to detect small CRISPR-Cas9 induced indels by agarose gel electrophoresis. Sci Rep. 2019;9(1):4437. doi: 10.1038/s41598-019-39950-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tveden-Nyborg P, Bergmann TK, Jessen N, Simonsen U, Lykkesfeldt J. BCPT policy for experimental and clinical studies. Basic Clin Pharmacol Toxicol. 2021;128(1):4–8. doi: 10.1111/bcpt.13492 [DOI] [PubMed] [Google Scholar]
  • 27.Shiu FH, Wong JC, Yamamoto T, et al. Mice lacking full length Adgrb1 (Bai1) exhibit social deficits, increased seizure susceptibility, and altered brain development. Exp Neurol. 2022;351:113994. doi: 10.1016/j.expneurol.2022.113994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wong JC, Grieco SF, Dutt K, et al. Autistic-like behavior, spontaneous seizures, and increased neuronal excitability in a Scn8a mouse model. Neuropsychopharmacology. 2021;46(11):2011–2020. doi: 10.1038/s41386-021-00985-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yang M, Crawley JN. Simple behavioral assessment of mouse olfaction. Curr Protoc Neurosci. 2009;Chapter 8:Unit 8 24;48 (1):Unit 8.24. doi: 10.1002/0471142301.ns0824s48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wong JC, Dutton SB, Collins SD, Schachter S, Escayg A. Huperzine A provides robust and sustained protection against induced seizures in Scn1a mutant mice. Front Pharmacol. 2016;7:357. doi: 10.3389/fphar.2016.00357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wong JC, Shapiro L, Thelin JT, et al. Nanoparticle encapsulated oxytocin increases resistance to induced seizures and restores social behavior in Scn1a-derived epilepsy. Neurobiol Dis. 2021;147:105147. doi: 10.1016/j.nbd.2020.105147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Giddens MM, Wong JC, Schroeder JP, et al. GPR37L1 modulates seizure susceptibility: evidence from mouse studies and analyses of a human GPR37L1 variant. Neurobiol Dis. 2017;106:181–190. doi: 10.1016/j.nbd.2017.07.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Shapiro L, Gado F, Manera C, Escayg A. Allosteric modulation of the cannabinoid 2 receptor confers seizure resistance in mice. Neuropharmacology. 2021;188:108448. doi: 10.1016/j.neuropharm.2021.108448 [DOI] [PubMed] [Google Scholar]
  • 34.Martin MS, Tang B, Papale LA, Yu FH, Catterall WA, Escayg A. The voltage-gated sodium channel Scn8a is a genetic modifier of severe myoclonic epilepsy of infancy. Hum Mol Genet. 2007;16(23):2892–2899. doi: 10.1093/hmg/ddm248 [DOI] [PubMed] [Google Scholar]
  • 35.Shapiro L, Wong JC, Escayg A. Reduced cannabinoid 2 receptor activity increases susceptibility to induced seizures in mice. Epilepsia. 2019;60(12):2359–2369. doi: 10.1111/epi.16388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Barton ME, Klein BD, Wolf HH, White HS. Pharmacological characterization of the 6 Hz psychomotor seizure model of partial epilepsy. Epilepsy Res. 2001;47(3):217–227. doi: 10.1016/s0920-1211(01)00302-3 [DOI] [PubMed] [Google Scholar]
  • 37.Krasowski MD. Differential modulatory actions of the volatile convulsant flurothyl and its anesthetic isomer at inhibitory ligand-gated ion channels. Neuropharmacology. 2000;39(7):1168–1183. doi: 10.1016/s0028-3908(99)00221-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liao HM, Chao YL, Huang AL, et al. Identification and characterization of three inherited genomic copy number variations associated with familial schizophrenia. Schizophr Res. 2012;139(1–3):229–236. doi: 10.1016/j.schres.2012.05.015 [DOI] [PubMed] [Google Scholar]
  • 39.Lips ES, Cornelisse LN, Toonen RF, et al. Functional gene group analysis identifies synaptic gene groups as risk factor for schizophrenia. Mol Psychiatry. 2012;17(10):996–1006. doi: 10.1038/mp.2011.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Heinzen EL, Radtke RA, Urban TJ, et al. Rare deletions at 16p13.11 predispose to a diverse spectrum of sporadic epilepsy syndromes. Am J Hum Genet. 2010;86(5):707–718. doi: 10.1016/j.ajhg.2010.03.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Scuderi C, Saccuzzo L, Vinci M, et al. Biallelic intragenic duplication in ADGRB3 (BAI3) gene associated with intellectual disability, cerebellar atrophy, and behavioral disorder. Eur J Hum Genet. 2019;27(4):594–602. doi: 10.1038/s41431-018-0321-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zhu D, Li C, Swanson AM, et al. BAI1 regulates spatial learning and synaptic plasticity in the hippocampus. J Clin Invest. 2015;125(4):1497–1508. doi: 10.1172/JCI74603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Okajima D, Kudo G, Yokota H. Antidepressant-like behavior in brain-specific angiogenesis inhibitor 2-deficient mice. J Physiol Sci. 2011;61(1):47–54. doi: 10.1007/s12576-010-0120-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hochreiter-Hufford AE, Lee CS, Kinchen JM, et al. Phosphatidylserine receptor BAI1 and apoptotic cells as new promoters of myoblast fusion. Nature. 2013;497(7448):263–267. doi: 10.1038/nature12135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Park D, Tosello-Trampont AC, Elliott MR, et al. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature. 2007;450(7168):430–434. doi: 10.1038/nature06329 [DOI] [PubMed] [Google Scholar]
  • 46.Amson R, Lassalle JM, Halley H, et al. Behavioral alterations associated with apoptosis and down-regulation of presenilin 1 in the brains of p53-deficient mice. Proc Natl Acad Sci U S a. 2000;97(10):5346–5350. doi: 10.1073/pnas.97.10.5346 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Engel T, Tanaka K, Jimenez-Mateos EM, Caballero-Caballero A, Prehn JH, Henshall DC. Loss of p53 results in protracted electrographic seizures and development of an aggravated epileptic phenotype following status epilepticus. Cell Death Dis. 2010;1(10):e79. doi: 10.1038/cddis.2010.55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lee JH, Kim JY, Noh S, et al. Astrocytes phagocytose adult hippocampal synapses for circuit homeostasis. Nature. 2021;590(7847):612–617. doi: 10.1038/s41586-020-03060-3 [DOI] [PubMed] [Google Scholar]
  • 49.Chu Y, Jin X, Parada I, et al. Enhanced synaptic connectivity and epilepsy in C1q knockout mice. Proc Natl Acad Sci U S A. 2010;107(17):7975–7980. doi: 10.1073/pnas.0913449107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Filipello F, Morini R, Corradini I, et al. The microglial innate immune receptor TREM2 is required for synapse elimination and normal brain connectivity. Immunity. 2018;48(5):979–991 e8. doi: 10.1016/j.immuni.2018.04.016 [DOI] [PubMed] [Google Scholar]
  • 51.Jeong BC, Kim MY, Lee JH, et al. Brain-specific angiogenesis inhibitor 2 regulates VEGF through GABP that acts as a transcriptional repressor. FEBS Lett. 2006;580(2):669–676. doi: 10.1016/j.febslet.2005.12.086 [DOI] [PubMed] [Google Scholar]
  • 52.Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA. Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci U S a. 2002;99(18):11946–11950. doi: 10.1073/pnas.182296499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chew KS, Fernandez DC, Hattar S, Sudhof TC, Martinelli DC. Anatomical and behavioral investigation of C1ql3 in the mouse suprachiasmatic nucleus. J Biol Rhythms. 2017;32(3):222–236. doi: 10.1177/0748730417704766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Qi Y, Xiong W, Yu S, et al. Deletion of C1ql1 causes hearing loss and abnormal auditory nerve fibers in the mouse cochlea. Front Cell Neurosci. 2021;15:713651. doi: 10.3389/fncel.2021.713651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Parag RP, Yamamoto T, Van Meir EG. Novel isoforms of adhesion G protein coupled receptor B1 (ADGRB1/BAI1) generated from an alternative promoter in intron 17. Authorea. 2023. doi: 10.22541/au.168207124.40138233/v1 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon request.

RESOURCES