Application of in Utero Electroporation of G-Protein Coupled Receptor (GPCR) Genes, for Subcellular Localization of Hardly Identifiable GPCR in Mouse Cerebral Cortex
Nam-Ho Kim, Seunghyuk Kim, Jae Seung Hong, Sung Ho Jeon, and Sung-Oh Huh
Abstract
Lysophosphatidic acid (LPA) is a lipid growth factor that exerts diverse biological effects through its cognate receptors (LPA1-LPA6). LPA1, which is predominantly expressed in the brain, plays a pivotal role in brain development. However, the role of LPA1 in neuronal migration has not yet been fully elucidated. Here, we delivered LPA1 to mouse cerebral cortex using in utero electroporation. We demonstrated that neuronal migration in the cerebral cortex was not affected by the overexpression of LPA1. Moreover, these results can be applied to the identification of the localization of LPA1. The subcellular localization of LPA1 was endogenously present in the perinuclear area, and overexpressed LPA1 was located in the plasma membrane. Furthermore, LPA1 in developing mouse cerebral cortex was mainly expressed in the ventricular zone and the cortical plate. In summary, the overexpression of LPA1 did not affect neuronal migration, and the protein expression of LPA1 was mainly located in the ventricular zone and cortical plate within the developing mouse cerebral cortex. These studies have provided information on the role of LPA1 in brain development and on the technical advantages of in utero electroporation.
INTRODUCTION
Lysophosphatidic acid (LPA; 1-acyl-sn-glycerol-3-phosphate), which is a phospholipid growth factor, exerts diverse biological effects on neuronal cells, including proliferation, differentiation, survival, morphological change, and migration (Choi et al., 2010; Ishii et al., 2004; Moolenaar et al., 2004; Yung et al., 2014). LPA signals through at least six specific membrane-bound G protein-coupled receptors (GPCRs) that are designated as LPA1/Vzg-1/Edg2, LPA2/Edg4, LPA3/Edg7, LPA4/p2y9/GPR23, LPA5/GPR92, and LPA6/p2y5 (An et al., 1998; Bandoh et al., 1999; Hecht et al., 1996; Kotarsky et al., 2006; Lee et al., 2006; Noguchi et al., 2003; Pasternack et al., 2008). The lysophosphatidic acid receptor-1 (LPA1), which was the first LPA receptor identified (Hecht et al., 1996), was found to be involved in LPA signaling in the development of the central nervous system through studies involving the targeted deletion of LPA1 (Contos et al., 2000; Estivill-Torrus et al., 2008; Matas-Rico et al., 2008). This receptor was shown to be expressed in the neurogenic region of the embryonic neocortical region, which is called the ventricular zone (VZ) (Hecht et al., 1996).
Following the identification of LPA1, studies were directed towards understanding the role of LPA in the cortical development of mice. When LPA was not present in the VZ, neural progenitor cells underwent proliferation, differentiation, and cell death. In the presence of LPA, cell rounding and retraction fiber formation were observed in mouse cortical primary cells (Fukushima et al., 2000). In addition, LPA signaling regulated the formation of cerebral cortical folds that resemble gyri by affecting proliferation, differentiation, and cell survival during embryonic development (Kingsbury et al., 2003). The mouse embryonic cerebral cortex exhibited gene expression profiles of two LPA receptors (LPA1 and LPA2), suggesting important roles of LPA1 and LPA2 in cerebral cortical development (Kingsbury et al., 2003). However, in many LPA studies, it has been difficult to localize the LPA receptor in virto.
Formerly, studies of the functional consequence and expression pattern of the LPA1 receptor were performed with in situ hybridization and reverse transcription polymerase chain reaction analysis (Cheng et al., 2009; Hecht et al., 1996; Kim et al., 2006; Spohr et al., 2008; Sun et al., 2010).
Since LPA1 receptors, like all G protein-couple receptors, cannot be easily visualized by an antibody, it is hard to discern the actual localization of the protein. Some papers have used LPA1 antibodies to show the subcellular localization and expression of LPA1. These studies examined antibody specificity by blocking the signal with the overexpression of antisense LPA receptors or LPA receptor peptides (Gobeil et al., 2003; Liszewska et al., 2009; Moughal et al., 2004; Waters et al., 2006; Zheng et al., 2001). However, these studies were not conclusive.
In this study, we ectopically overexpressed LPA1 in the developing mouse cerebral cortex using in utero electroporation. LPA1 overexpressed mice did not show obvious abnormalities in cerebral cortical development. In addition, we examined the subcellular localization and tissue distribution of LPA1 in embryonic brain. The subcellular localization of LPA1 in LPA1-overexpressing cells was mainly located in the plasma membrane and endogenously located in the perinuclear area. The tissue distribution of LPA1 was mainly observed in the VZ and cortical plate (CP) of the embryonic cortex. Our studies revealed the subcellular localization of LPA1 áå îáîç and showed the technical advantage of antibody validation using in utero electroporation.
MATERIALS AND METHODS
Animal
Pregnant mice of C57BL/6N strain were purchased from Orient Bio (Korea). Stage E0.5 was defined as noon on the day of the vaginal plug. Fetuses at E13.5, E15.5, and E17.5 were used for experiments.
Materials
All chemicals used were of analytical grade if not stated otherwise. Antibody to β-actin was obtained from Cell Signaling Technology (USA). Antibodies to LPA1, and green fluorescent protein (GFP) were purchased from Abcam (UK). Dulbecco’s modified Eagle’s Medium (DMEM), Opti-MEM I reduced-serum medium, 100 unit penicillin/100 μg streptomycin, fetal bovine serum (FBS), lipofectamine 2000, and DAPI were obtained from Invitrogen (USA).
Cell culture
TR cells which is derived from neocortical neuroblast cells infected with the oncogenes Large T and vras (Chun and Jaenisch, 1996) were maintained as monolayer cultures in Opti-MEM I reduced-serum medium supplemented with 2.5% heat-inactivated fetal bovine serum, 20 mM glucose, 55 μM 2-mercaptoethanol, and 100 unit penicillin/100 μg streptomycin. B103 rat neuroblastoma cells were maintained in DMEM supplemented with 10% FBS. B103 cells were transiently transfected with pCAGIG or pCAGIG-LPA1 expression plasmid using the Lipofectamine 2000 reagent. After 24 h, the fluorescent images were acquired with an inverted microscope (IX70; Olympus).
Isolation of total protein and Western blot analysis
TR cells were lysed in lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM ethylene glycol tetraacetic acid (EGTA), 1 mM ethylenediaminetetraacetic acid (EDTA), 1% Triton X-100, 1 mM Na3VO4, 5 mM NaF, and protease inhibitor cocktail). After incubation on ice for 30 min, the lysates were centrifuged (15,000 × g, 15 min). Supernatants were collected and protein concentrations were determined by Bradford assay (Bio-Rad, USA). Equal amounts of protein were boiled and separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) transferred to polyvinylidene difluoride membranes (Millipore, USA), and blocked with 5% non-fat milk. Membranes were incubated in primary antibody overnight at 4°C. Membranes were then washed in TBST (10 mM Tris, 140 mM NaCl, 0.1% Tween 20, pH 7.6), incubated with appropriate secondary antibody, and washed again in TBST. Bands were visualized by chemiluminescence and exposed to X-ray film.
DNA constructs
pCAGIG-LPA1 expression vector design was based on the pCAGIG vector. The pCAGIG vector, which contains the IRES-EGFP cDNA under the control of the CMV enhancer and chick β-actin promoter, was a gift from Dr. C. Cepko (Matsuda and Cepko, 2004). The full coding sequence for murine LPA1 was obtained by reverse transcription PCR. Total RNA was prepared from the cerebellum of adult C57BL/6N mice using TRIzol reagent (Invitrogen). Total RNA (2 μg) was converted to cDNA using AMV reverse transcriptase (Promega, USA). The coding regions of mouse LPA1 were PCR amplified from cDNA with the following 5′-EcoRI-mLPA1 and 3′-NotI-mLPA1 primer sets: 5′-CCGGAATTCATGGCAGCTGCCTCTACTT-3′, and 5′-GAGAGCGGCCGCTACACGGTCACCCCAGA-3′. The PCR product was subcloned into the pCAGIG vector at EcoRI and NotI sites. pCAGIG-LPA1 construct was confirmed by automated sequencing.
In utero electroporation
Timed-pregnant C57BL/6N females were anesthetized at stage embryonic day 13.5 (E13.5) with isoflurane (4% during induction, 2.5% during surgery), and the uterine horns were exposed by way of a laparotomy. The 1 μl of expression vector (4 μg/μl for pCAGIG and pCAGIG-LPA1 constructs) in phosphate buffered saline (PBS) containing 0.05% fast green (Sigma-Aldrich, USA) was injected into the lateral ventricle of the embryo using a glass capillary with a length of 90 mm and a diameter of 1 mm (GD-1; Narishige, Japan). Electroporation was performed with a Tweezertrodes (diameter, 5 mm; BTX, USA) with 5 pulses of 45 V for 50 millisecond duration and 950 millisecond interval using a square-wave pulse generator (ECM 830; BTX). The uterine horns were then returned into the abdominal cavity, the wall and skin were sutured, and embryos were allowed to continue their normal development.
Immunohistochemistry
Electroporated embryonic brains were fixed with 4% paraformaldehyde (PFA) at 4°C for 2 h. Brains were cryoprotected in 30% sucrose/1× PBS at 4°C overnight and embedded in Tissue-Tek OCT (Sakura Finetek, USA). Cryosections (10 μm) were collected on MAS-coated glass slides (Matsunami Glass, Japan). Sections on glass slides were treated with heat in citrate buffer (10 mM, pH 6.0) at 95°C for 5 min. Samples were blocked in PBST containing 10% normal goat serum. The sections were incubated with primary antibodies against GFP (Abcam) (1:1000) and LPA1 (Abcam) (1:1000) overnight at 4°C and then incubated in Alexa 488 and Alexa 568 conjugated secondary antibodies (Invitrogen) (1:1000) for one hour at room temperature. After washing, the specimens were mounted onto cover slips using Vectashield (Vector Laboratories, USA). The fluorescent images were acquired with a laser scanning confocal microscope (LSM510; Zeiss). For identify distribution of the GFP positive cells, images were converted gray values and normalized to background staining. Images were divide 10 equal bins spanning the cortical thickness and measured by ImageJ program (NIH).
In situ hybridization
Cryosections (18 μm) were collected on MAS-coated glass slides (Matsunami Glass). mRNA for LPA1 was detected by in situ hybridization using digoxygenin (DIG)-labeled antisense riboprobes. The sections were treated with proteinase K (1 μg/ml, 5–30 min, room temperature) and hybridized with 0.3 μg/ml riboprobe in a hybridization buffer (50% formamide, 20 mM Tris-HCl at pH 7.5, 600 mM NaCl, 1 mM EDTA, 10% dextran sulfate, 200 μg/ml yeast tRNA, 1× Denhardt’s solution, 0.25% SDS) at 65°C overnight. The sections were washed three times with 1× SSC containing 50% formamide at 65°C, followed by maleic acid buffer (0.1 M, pH 7.5) containing 0.1% Tween 20 and 0.15 M NaCl. DIG-labeled probe was visualized by overnight incubation of the sections with anti-DIG antibody conjugated to alkaline phosphatase (1:2,000; Roche, USA) and NBT/BCIP reaction. The open reading frame of Lpar1 antisense riboprobe was synthesized using a digoxygenin-labeled riboprobe with T7 RNA polymerase and a DIG-RNA labeling mix according to the manufacturer (Roche). The Lpar1 riboprobe was a gift from Dr. Jerold Chun (The Scripps Research Institute).
RESULTS
Characterization of the spatiotemporal-specific LPA1-overexpressing mouse cerebral cortex
To evaluate the importance of LPA1 in neuronal migration in the developing cerebral cortex, we examined the overexpression of LPA1 in the VZ by in utero electroporation. First, we made a vector containing LPA1 and tested it in B103 rat neuroblastoma cells. pGAGIG-LPA1 encoding [LPA1/enhanced green fluorescent protein (EGFP)] - transfected cells displayed a flattened and more migratory morphology compared with pCAGIG (encoding EGFP)-transfected cells (data not shown). We injected a construct encoding EGFP or LPA1/EGFP into the lateral ventricle of E13.5 mouse embryos and transferred it into neuronal progenitor cells in the VZ by in utero electroporation (Saito, 2006). After allowing normal in vivo embryonic development, immunohistochemistry for GFP was performed on coronal brain sections obtained from E15.5 mice. The expression of EGFP was detectable in many neurons of mice transfected with EGFP (Fig. 1A) or with LPA1 and EGFP (Fig. 1B). In EGFP-transfected control (Figs. 1A and 1C), transfected cells were mainly located in the subventricular zone (SVZ) and intermediate zone (IZ), and some populations entered the cortical plate (CP). These neuronal migration patterns correlated with previously reported results (Langevin et al., 2007). With LPA1/EGFP-transfection (Figs. 1B and 1D), LPA1 did not affect neuronal migration or neuronal morphology in developing mouse cerebral cortex. Although LPA1 overexpression appeared to slightly delay neuronal migration in CP, it was not statistically significant. (Figs. 1C and 1D). These results indicate that LPA1 overexpression in cerebral cortex did not affect radial migration of neuronal progenitor cells.
Subcellular localization of LPA1 in LPA1-overexpressing mice
To delineate the subcellular localization of LPA1, we examined its protein distribution in EGFP- and LPA1/EGFP-transfected mice cerebral cortex. First, we tested the antibody specificity to LPA1. Western blot analysis of EGFP and LPA1/EGFP overexpressing TR neocortical neuroblast cells, which revealed a single band at the expected size for LPA1 (Fig. 2), demonstrated the specificity of the antibody used to detect LPA1. We next characterized the subcellular localization of LPA1 using immunohistochemistry (Fig. 3). At E15.5, 2 days after in utero electroporation, we performed GFP and LPA1 double immunostaining in EGFP- and LPA1/EGFP-transfected mice cerebral cortex. In EGFP-transfected controls, LPA1 did not co-localize with the GFP signal (Figs. 3A–3C). Interestingly, endogenous LPA1-positive signal, which was observed in the nuclear/perinuclear area of neuronal cells located in the VZ, formed a spot-like pattern (Fig. 3D). In LPA1/EGFP-overexpressing cortices, LPA1 expression co-localized with GFP (Figs. 3E–3G). As shown in Fig. 3H, LPA1 was mainly located in plasma membrane in LPA1-overexpressing neuronal cells. Additionally, endogenous LPA1-positive signals were the same as those seen in EGFP transfected controls (Fig. 3G). Thus, these data show that the endogenous subcellular localization of LPA1, which exhibited a spot-like pattern, was located in the nuclear/perinuclear area, whereas the LPA1 in LPA1-overexpressing cells was located in the plasma membrane.

LPA1 is mainly expressed in ventricular zone of mouse embryonic cerebral cortex
Due to the fact that our Western blotting and in utero electroporation experiment revealed that the LPA1 antibody was specific for detecting LPA1, we further identified the LPA1 protein expression pattern in the cerebral cortex of (E13.5 to E17.5) embryonic mice (Figs. 5 and 6). First, we identified that the Lpar1 transcript was mainly expressed in the VZ of cerebral cortex using in situ hybridization, as previously described (Hecht et al., 1996) (Fig. 4). At E13.5, Lpar1 antisense riboprobes revealed that a high level of expression of Lpar1 was detected strictly in the VZ of cerebral cortex (Fig. 4A). At E15.5, the level of Lpar1 mRNA expression, which were exhibited in the VZ were slightly diminished (Fig. 4C). At E17.5, Lpar1 mRNA in VZ was barely detectable (Fig. 4E). Immunohistochemical analysis confirmed that LPA1 was located in the cerebral cortex at E13.5 (Fig. 5). In particular, LPA1 levels were highly enriched within the VZ (Figs. 5A and 5B) and the lens (Figs. 5A and 5C). To examine the expression pattern of LPA1 during cortical development, we performed immunostaining on sagittal brain sections obtained from E13.5 to E17.5 mice (Fig. 6). At E13.5, immunostaining of LPA1 revealed that a high level of LPA1 was detected in the VZ of cerebral cortex. At E15.5, LPA1 levels were high in the VZ and the CP. At E17.5, the levels of LPA1 were highly enriched within the VZ and upper layer of the CP. Taken together, these data demonstrate that LPA1 was expressed in the VZ and upper layer of the CP in the cerebral cortex of developing mice.


DISCUSSION
The aim of the present study was to characterize functional consequence of LPA1 in the developing cerebral cortex. Our results provide an additional understanding of the role of LPA1 in the developing cerebral cortex and of the technical application of in utero electroporation for the validation of antibodies.
We used in utero electroporation to uncover the role of LPA1 in neuronal migration from the VZ to the CP. The overexpression of LPA1 in newly postmitotic cells of the VZ did not show any obvious abnormality in the neuronal migration to the CP (Fig. 1). LPA1, which is endogenously expressed in the VZ (Hecht et al., 1996), has an important role in the maintenance of neuroprogenitor pools in the VZ (Kingsbury et al., 2003). The LPA receptor-mediated signaling is regulated by their ligand, LPA. Thus, the levels of LPA may be low in the SVZ and IZ. Thus, the ectopic expression of LPA1 did not change neuronal migration in the cerebral cortex.
An examination of the subcellular localization of LPA1 was performed by immunohistochemistry (Fig. 3). In LPA1-overexpressing cerebral cortical cells, LPA1, which colocalized with EGFP fluorescence, was predominantly expressed in the plasma membrane (Fig. 3H). These results were similar to those previously reported in a study of LPA1 overexpression in a cell-based assay (Avendano-Vazquez et al., 2005; Murph et al., 2003; Urs et al., 2005).
In EGFP expressing control cerebral cortex, LPA1 and EGFP signal did not colocalize, and the endogenous LPA1-positive signal was located in the perinuclear area and looked like an LPA1 oligomer (Fig. 3D). G protein-coupled receptor (GPCR) oligomerization has recently been widely accepted, and the number of documented oligomeric GPCR combinations is extensive (Filizola, 2010; Gurevich and Gurevich, 2008; Maggio et al., 2005; Milligan, 2009; Vidi et al., 2011). Active GPCRs are specifically phosphorylated by G-protein-coupled receptor kinases (GRKs), initiating arrestin recruitment. Receptor/arrestin complexes than recruit two components of the internalization machinery (clathrin and AP-2) and a variety of other proteins, initiating the second round of signaling (DeWire et al., 2007; Moore et al., 2007). Thus, GPCR oligomerization may be regulated by arrestin-mediated signaling (Gurevich and Gurevich, 2008). Arrestin is important in the LPA induced signaling cascade, and LPA1 internalization is regulated by arrestin mediated pathway (DeWire et al., 2007; Gesty-Palmer et al., 2005; Sun and Lin, 2008; Sun and Yang, 2010; Urs et al., 2005; 2008). Another report showed that LPA1 can dimerize (Zaslavsky et al., 2006). Our observation of ventricular zone cells that were immunopositive to LPA1 antibody with puncta morphology is intriguing. We speculate that these intracellular puncta might have been formed after LPA1 are bound to ligands (i.e., lysophosphatitic acids), followed by internalization of this receptor-ligand complex into the cells. Taken together, these results suggest that activated LPA1 is internalized and oligomerized by arrestin scaffolding.
In a previous in situ hybridization study, the Lpar1 expression pattern was mainly located in the VZ of the cerebral cortex of developing mouse (Hecht et al., 1996). We performed in situ hybridization of Lpar1 in the cerebral cortex and found the same expression pattern of the Lpar1 transcript (Fig. 4). In Figs. 5 and 6, the protein expression pattern of LPA1 is mainly located in the VZ and the CP. During cortical development, postmitotic neurons migrate toward the CP from the VZ and pass the IZ. Within the CP, radially migrating cells become arranged in an inside-out pattern in which the earlier generated neurons occupy deeper layers, and those generated later become located in more superficial layers (Aboitiz et al., 2001). These previous reports indicated that the transcripts of Lpar1 are located in the VZ, and LPA1-expressing cells migrate toward the CP from VZ. Thus, LPA1 protein is located in the VZ and CP of the cerebral cortex of mice. This observation requires further studies with functional consequence of LPA1 expressed in VZ and CP.
Nowadays, in utero electroporation is a widely used and well established experimental method. It has a several advantage like region-specific, cell type-specific, inducible, and multiple gene targeting. Moreover, it can apply to region dependent behaviors and functional outcome test (De Vry et al., 2010; Taniguchi et al., 2012). Besides the above advantages of the in utero electroporation, it can easily introduce specific genes in vivo condition. Despite many targeted GPCR antibody is available but can’t easily find validated antibody, for this reason in utero electroporation is a powerful tools for quick and easily find good antibody from hardly discern GPCR.
In summary, our results provide information on the subcellular localization and tissue distribution of LPA1 and the technical advantages of the use of in utero electroporation for the validation of antibodies.
Article information
Articles from Mol. Cells are provided here courtesy of Mol. Cells
References
- Aboitiz, F, Morales, D, and Montiel, J (2001). The inverted neurogenetic gradient of the mammalian isocortex: development and evolution. Brain Res Brain Res Rev. 38, 129-139.
- An, S, Bleu, T, Hallmark, OG, and Goetzl, EJ (1998). Characterization of a novel subtype of human G protein-coupled receptor for lysophosphatidic acid. J Biol Chem. 273, 7906-7910.
- Avendano-Vazquez, SE, Garcia-Caballero, A, and Garcia-Sainz, JA (2005). Phosphorylation and desensitization of the lysophosphatidic acid receptor LPA1. Biochem J. 385, 677-684.
- Bandoh, K, Aoki, J, Hosono, H, Kobayashi, S, Kobayashi, T, Murakami-Murofushi, K, Tsujimoto, M, Arai, H, and Inoue, K (1999). Molecular cloning and characterization of a novel human G-protein-coupled receptor, EDG7, for lysophosphatidic acid. J Biol Chem. 274, 27776-27785.
- Cheng, Y, Makarova, N, Tsukahara, R, Guo, H, Shuyu, E, Farrar, P, Balazs, L, Zhang, C, and Tigyi, G (2009). Lysophosphatidic acid-induced arterial wall remodeling: requirement of PPAR-gamma but not LPA1 or LPA2 GPCR. Cell Signal. 21, 1874-1884.
- Choi, JW, Herr, DR, Noguchi, K, Yung, YC, Lee, CW, Mutoh, T, Lin, ME, Teo, ST, Park, KE, and Mosley, AN (2010). LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol. 50, 157-186.
- Chun, J, and Jaenisch, R (1996). Clonal cell lines produced by infection of neocortical neuroblasts using multiple oncogenes transduced by retroviruses. Mol Cell Neurosci. 7, 304-321.
- Contos, JJ, Fukushima, N, Weiner, JA, Kaushal, D, and Chun, J (2000). Requirement for the lpA1 lysophosphatidic acid receptor gene in normal suckling behavior. Proc Natl Acad Sci USA. 97, 13384-13389.
- De Vry, J, Martinez-Martinez, P, Losen, M, Temel, Y, Steckler, T, Steinbusch, HW, De Baets, MH, and Prickaerts, J (2010). In vivo electroporation of the central nervous system: a non-viral approach for targeted gene delivery. Prog Neurobiol. 92, 227-244.
- DeWire, SM, Ahn, S, Lefkowitz, RJ, and Shenoy, SK (2007). Beta- arrestins and cell signaling. Annu Rev Physiol. 69, 483-510.
- Estivill-Torrus, G, Llebrez-Zayas, P, Matas-Rico, E, Santin, L, Pedraza, C, De Diego, I, Del Arco, I, Fernandez-Llebrez, P, Chun, J, and De Fonseca, FR (2008). Absence of LPA1 signaling results in defective cortical development. Cereb Cortex. 18, 938-950.
- Filizola, M (2010). Increasingly accurate dynamic molecular models of G-protein coupled receptor oligomers: Panacea or Pandora’s box for novel drug discovery?. Life Sci. 86, 590-597.
- Fukushima, N, Weiner, JA, and Chun, J (2000). Lysophosphatidic acid (LPA) is a novel extracellular regulator of cortical neuroblast morphology. Dev Biol. 228, 6-18.
- Gesty-Palmer, D, El Shewy, H, Kohout, TA, and Luttrell, LM (2005). beta-Arrestin 2 expression determines the transcriptional response to lysophosphatidic acid stimulation in murine embryo fibroblasts. J Biol Chem. 280, 32157-32167.
- Gobeil, F, Bernier, SG, Vazquez-Tello, A, Brault, S, Beauchamp, MH, Quiniou, C, Marrache, AM, Checchin, D, Sennlaub, F, and Hou, X (2003). Modulation of pro-inflammatory gene expression by nuclear lysophosphatidic acid receptor type-1. J Biol Chem. 278, 38875-38883.
- Gurevich, VV, and Gurevich, EV (2008). GPCR monomers and oligomers: it takes all kinds. Trends Neurosci. 31, 74-81.
- Hecht, JH, Weiner, JA, Post, SR, and Chun, J (1996). Ventricular zone gene-1 (vzg-1) encodes a lysophosphatidic acid receptor expressed in neurogenic regions of the developing cerebral cortex. J Cell Biol. 135, 1071-1083.
- Ishii, I, Fukushima, N, Ye, X, and Chun, J (2004). Lysophospholipid receptors: signaling and biology. Annu Rev Biochem. 73, 321-354.
- Kim, J, Keys, JR, and Eckhart, AD (2006). Vascular smooth muscle migration and proliferation in response to lysophosphatidic acid (LPA) is mediated by LPA receptors coupling to Gq. Cell Signal. 18, 1695-1701.
- Kingsbury, MA, Rehen, SK, Contos, JJ, Higgins, CM, and Chun, J (2003). Non-proliferative effects of lysophosphatidic acid enhance cortical growth and folding. Nat Neurosci. 6, 1292-1299.
- Kotarsky, K, Boketoft, A, Bristulf, J, Nilsson, NE, Norberg, A, Hansson, S, Owman, C, Sillard, R, Leeb-Lundberg, LM, and Olde, B (2006). Lysophosphatidic acid binds to and activates GPR92, a G protein-coupled receptor highly expressed in gastrointestinal lymphocytes. J Pharmacol Exp Ther. 318, 619-628.
- Langevin, LM, Mattar, P, Scardigli, R, Roussigne, M, Logan, C, Blader, P, and Schuurmans, C (2007). Validating in utero electroporation for the rapid analysis of gene regulatory elements in the murine telencephalon. Dev Dyn. 236, 1273-1286.
- Lee, CW, Rivera, R, Gardell, S, Dubin, AE, and Chun, J (2006). GPR92 as a new G12/13- and Gq-coupled lysophosphatidic acid receptor that increases cAMP, LPA5. J Biol Chem. 281, 23589-23597.
- Liszewska, E, Reinaud, P, Billon-Denis, E, Dubois, O, Robin, P, and Charpigny, G (2009). Lysophosphatidic acid signaling during embryo development in sheep: involvement in prostaglandin synthesis. Endocrinology. 150, 422-434.
- Maggio, R, Novi, F, Scarselli, M, and Corsini, GU (2005). The impact of G-protein-coupled receptor hetero-oligomerization on function and pharmacology. FEBS J. 272, 2939-2946.
- Matas-Rico, E, Garcia-Diaz, B, Llebrez-Zayas, P, Lopez-Barroso, D, Santin, L, Pedraza, C, Smith-Fernandez, A, Fernandez-Llebrez, P, Tellez, T, and Redondo, M (2008). Deletion of lysophosphatidic acid receptor LPA1 reduces neurogenesis in the mouse dentate gyrus. Mol Cell Neurosci. 39, 342-355.
- Matsuda, T, and Cepko, CL (2004). Electroporation and RNA interference in the rodent retina in vivo and in vitro. Proc. Natl. Acad. Sci. USA. 101, 16-22.
- Milligan, G (2009). G protein-coupled receptor hetero-dimerization: contribution to pharmacology and function. Br J Pharmacol. 158, 5-14.
- Moolenaar, WH, van Meeteren, LA, and Giepmans, BN (2004). The ins and outs of lysophosphatidic acid signaling. Bioessays. 26, 870-881.
- Moore, CA, Milano, SK, and Benovic, JL (2007). Regulation of receptor trafficking by GRKs and arrestins. Annu Rev Physiol. 69, 451-482.
- Moughal, NA, Waters, C, Sambi, B, Pyne, S, and Pyne, NJ (2004). Nerve growth factor signaling involves interaction between the Trk A receptor and lysophosphatidate receptor 1 systems: nuclear translocation of the lysophosphatidate receptor 1 and Trk A receptors in pheochromocytoma 12 cells. Cell Signal. 16, 127-136.
- Murph, MM, Scaccia, LA, Volpicelli, LA, and Radhakrishna, H (2003). Agonist-induced endocytosis of lysophosphatidic acid-coupled LPA1/EDG-2 receptors via a dynamin2- and Rab5-dependent pathway. J Cell Sci. 116, 1969-1980.
- Noguchi, K, Ishii, S, and Shimizu, T (2003). Identification of p2y9/GPR23 as a novel G protein-coupled receptor for lysophosphatidic acid, structurally distant from the Edg family. J Biol Chem. 278, 25600-25606.
- Pasternack, SM, von Kugelgen, I, Aboud, KA, Lee, YA, Ruschendorf, F, Voss, K, Hillmer, AM, Molderings, GJ, Franz, T, and Ramirez, A (2008). G protein-coupled receptor P2Y5 and its ligand LPA are involved in maintenance of human hair growth. Nat Genet. 40, 329-334.
- Saito, T (2006). In vivo electroporation in the embryonic mouse central nervous system. Nat Protoc. 1, 1552-1558.
- Spohr, TC, Choi, JW, Gardell, SE, Herr, DR, Rehen, SK, Gomes, FC, and Chun, J (2008). Lysophosphatidic acid receptor-dependent secondary effects via astrocytes promote neuronal differentiation. J Biol Chem. 283, 7470-7479.
- Sun, J, and Lin, X (2008). Beta-arrestin 2 is required for lysophosphatidic acid-induced NF-kappaB activation. Proc Natl Acad Sci USA. 105, 17085-17090.
- Sun, W, and Yang, J (2010). Molecular basis of lysophosphatidic acid-induced NF-kappaB activation. Cell Signal. 22, 1799-1803.
- Sun, Y, Nam, JS, Han, DH, Kim, NH, Choi, HK, Lee, JK, Rhee, HJ, and Huh, SO (2010). Lysophosphatidic acid induces upregulation of Mcl-1 and protects apoptosis in a PTX-dependent manner in H19-7 cells. Cell Signal. 22, 484-494.
- Taniguchi, Y, Young-Pearse, T, Sawa, A, and Kamiya, A (2012). In utero electroporation as a tool for genetic manipulation in vivo to study psychiatric disorders: from genes to circuits and behaviors. Neuroscientist. 18, 169-179.
- Urs, NM, Jones, KT, Salo, PD, Severin, JE, Trejo, J, and Radhakrishna, H (2005). A requirement for membrane cholesterol in the beta-arrestin- and clathrin-dependent endocytosis of LPA1 lysophosphatidic acid receptors. J Cell Sci. 118, 5291-5304.
- Urs, NM, Kowalczyk, AP, and Radhakrishna, H (2008). Different mechanisms regulate lysophosphatidic acid (LPA)-dependent versus phorbol ester-dependent internalization of the LPA1 receptor. J Biol Chem. 283, 5249-5257.
- Vidi, PA, Ejendal, KF, Przybyla, JA, and Watts, VJ (2011). Fluorescent protein complementation assays: new tools to study G protein-coupled receptor oligomerization and GPCR-mediated signaling. Mol Cell Endocrinol. 331, 185-193.
- Waters, CM, Saatian, B, Moughal, NA, Zhao, Y, Tigyi, G, Natarajan, V, Pyne, S, and Pyne, NJ (2006). Integrin signalling regulates the nuclear localization and function of the lysophosphatidic acid receptor-1 (LPA1) in mammalian cells. Biochem J. 398, 55-62.
- Yung, YC, Stoddard, NC, and Chun, J (2014). LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res. 55, 1192-1214.
- Zaslavsky, A, Singh, LS, Tan, H, Ding, H, Liang, Z, and Xu, Y (2006). Homo- and hetero-dimerization of LPA/S1P receptors, OGR1 and GPR4. Biochim. Biophys. Acta. 1761, 1200-1212.
- Zheng, Y, Kong, Y, and Goetzl, EJ (2001). Lysophosphatidic acid receptor-selective effects on Jurkat T cell migration through a Matrigel model basement membrane. J Immunol. 166, 2317-2322.