Next Article in Journal
Do Changes in ACE-2 Expression Affect SARS-CoV-2 Virulence and Related Complications: A Closer Look into Membrane-Bound and Soluble Forms
Next Article in Special Issue
ERα36–GPER1 Collaboration Inhibits TLR4/NFκB-Induced Pro-Inflammatory Activity in Breast Cancer Cells
Previous Article in Journal
Renin-Angiotensin System in Pathogenesis of Atherosclerosis and Treatment of CVD
Previous Article in Special Issue
New Structural Perspectives in G Protein-Coupled Receptor-Mediated Src Family Kinase Activation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The LPA3 Receptor: Regulation and Activation of Signaling Pathways

Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México CP 04510, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(13), 6704; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136704
Submission received: 24 May 2021 / Revised: 8 June 2021 / Accepted: 12 June 2021 / Published: 23 June 2021

Abstract

:
The lysophosphatidic acid 3 receptor (LPA3) participates in different physiological actions and in the pathogenesis of many diseases through the activation of different signal pathways. Knowledge of the regulation of the function of the LPA3 receptor is a crucial element for defining its roles in health and disease. This review describes what is known about the signaling pathways activated in terms of its various actions. Next, we review knowledge on the structure of the LPA3 receptor, the domains found, and the roles that the latter might play in ligand recognition, signaling, and cellular localization. Currently, there is some information on the action of LPA3 in different cells and whole organisms, but very little is known about the regulation of its function. Areas in which there is a gap in our knowledge are indicated in order to further stimulate experimental work on this receptor and on other members of the LPA receptor family. We are convinced that knowledge on how this receptor is activated, the signaling pathways employed and how the receptor internalization and desensitization are controlled will help design new therapeutic interventions for treating diseases in which the LPA3 receptor is implicated.

1. Introduction

Lysophosphatidic acid (LPA) is a simple lipid comprising a phosphate group and a fatty acid, joined by ester bonds to a glycerol moiety, which is considered the backbone of this molecule [1,2] (Figure 1). LPA has a wide distribution in the organism. It is found in tissues and fluids, likely due to its chemical and physical characteristics, particularly its low molecular weight and water solubility [3].
Two pathways synthesize LPA. In the intracellular pathway, phospholipids (phosphatidylcholine, phosphatidylserine, or phosphatidylethanolamine) or diacylglycerol are the metabolic precursors of LPA through the action of phospholipase D or diacylglycerol kinase, respectively. These enzymes promote the synthesis of phosphatidic acid, which is converted into LPA through catalysis by cytoplasmic lysophospholipases A1 or A2 [4,5]. Other molecules from which LPA is synthesized include glycerol-3-phosphate and monoacyl-glycerol. In these processes, we find the participation of the enzymes glycerophosphate acyltransferase and monoacyl-glycerol kinase, respectively [3,6,7].
In the extracellular pathway, LPA is generated from lysophosphatidylcholine, which is found in the extracellular leaflet of plasma membranes or bound to proteins (such as albumin). In this case, secreted lysophospholipases A1 or A2 split a fatty acid from phosphatidylcholine, synthesizing lysophosphatidylcholine, and then converting it into LPA by a phospholipase D, generally denominated Autotaxin [6,8,9].
LPA is degraded by various enzymes, including LPA acyltransferase, which transfers an acyl group from acyl-CoA to LPA, generating phosphatidic acid; LPA lipid phosphatase, which can remove the phosphate group from LPA, generating monoacylglycerol, and lysophospholipases, which lead to the hydrolysis of the acyl group of LPA, producing a free fatty acid and glycerol 3-phosphate [1,2].
LPA is considered a “bioactive lipid”, implying that it, in addition to its role in phospholipid metabolism, regulates a diverse range of cellular and organism responses such as angiogenesis [8,10,11], neuritic retraction [12,13,14], cell migration [15,16], cell proliferation [17,18,19], reorganization of the cytoskeleton [10,20], development of the central nervous system [8,20,21], neuronal myelination [20,22], pain [23,24], obesity [25], and cancer [26,27,28,29], among many others. These functions are performed by LPA through the activation of six receptors [5,30,31,32,33,34,35].
These receptors are called lysophosphatidic or LPA receptors and are classified into two families. The first family is the lysophospholipid family of receptors, related to those for other phospholipids and including the LPA1, LPA2, and LPA3 receptors. The second family is phylogenetically related to the purinergic receptors and includes the LPA4, LPA5, and LPA6 receptors [1,3,8,36].
These LPA receptors belong to the G protein-coupled receptor (GPCR) superfamily. They are structurally constituted of seven transmembrane hydrophobic domains connected by three intracellular loops and three extracellular loops, with an extracellular amino-terminal group and an intracellular carboxyl terminus. According to the classification criteria in the GRAFS (Glutamate, Rhodopsin, Adhesion, Frizzled/Taste2, and Secretin groups) system [37] and in the AF system, all of these receptors belong to family A [37,38,39]. These receptors are associated for their signaling with heterotrimeric GTPases or “G” proteins. LPA receptors can activate different Gα proteins (Gαq/11i/o, Gα12/13, Gαs); some of these receptors are considered promiscuous because they can activate different G proteins and downstream signaling pathways that regulate various physiological functions as well as being involved in the pathogenesis of different diseases [5,8,39] (Figure 2).
The activation of GPCRs by their agonists leads to conformational changes promoting heterotrimeric G protein interaction and the exchange of GDP for GTP in their Gα subunits, favoring the dissociation of these heterotrimeric proteins into their Gα subunits, and the βγ complexes, which separately mediate the activation of downstream proteins [8,40,41]. The termination/attenuation of signaling is associated with receptor phosphorylation by different protein kinases (including G protein-coupled receptor kinases (GRKs) and second messenger-activated kinases, among others) [42,43,44,45,46,47,48,49,50]. Such phosphorylations facilitate interaction with β-arrestins, disfavoring receptor-G protein interaction (therefore, decreasing G protein-mediated signaling), recruiting the endocytic machinery, promoting receptor internalization (Figure 3), and activating alternative signaling processes [49,50,51,52,53].
As indicated, the LPA receptors belonging to the lysophospholipid family include the LPA1-3 receptors. These receptors have been studied in more detail (reviewed in [8,9]). The LPA1 receptor is a 364 amino acid protein, which interacts mainly with Gi/o, Gq/11, and G12/13. In mice, knocking out the expression of this receptor subtype markedly affects the development of the central nervous system and decreases survival (50% perinatal death). Alteration of LPA1 expression has been associated with cancer, neuropathic pain and fibrosis of the lungs. LPA2 is a protein of 348 amino acid residues that interacts with Gi/o, Gq/11, and G12/13. Constitutive receptor loss in mice produces an essentially normal phenotype; however, this receptor contributes to the development and function of synapsis in embryos and adult mice. It has also been associated with some types of cancer and lung functional alterations, such as asthma. The LPA3 receptor is a GPCR whose activation mainly promotes the recruitment of two G proteins: Gαq/11 and Gαi/o; therefore, it is considered promiscuous. The LPA3 receptor regulates different signal pathways, as depicted in Figure 4. It should be mentioned that LPA receptors (LPA1-3 form homo- and hetero-dimers within the subgroup and hetero-dimers with other receptors such as those of the sphingosine 1-phosphate receptor (S1P1–3) and the proton-sensing GPCR, GPR4 [54]. This adds a new level of complexity in signaling and regulation, which we consider important to mention, but it is not considered in the present review.
This review focuses on lysophosphatidic acid receptor 3 (LPA3), which participates in different functions which will be briefly described and discussed, and on the information available on the structure of this receptor and its regulation.

2. The LPA3 Receptor Is Involved in Physiology and Pathology

2.1. Antioxidant Enzymes Are Regulated by LPA3

LPA3 activation appears to play a critical role in regulating the expression of the enzymes that eliminate reactive oxygen species (ROS), primarily through transcription factor NF-E-p45-related factor 2 (Nrf2). Activation of this factor promotes the expression of enzymes such as superoxide dismutase (SOD), glutathione peroxidase 1 (GPx1), heme oxygenase-1 (HO-1), and NAD(P)H: quinone acceptor oxidoreductase (NQO1) [14,55,56]. These enzymes protect cells from the damage produced by the inflammatory process at the beginning of many diseases.
Chen and coworkers [55] showed that LPA3 activation increases the expression of antioxidant enzymes in a model of progeria syndrome. In this study, the authors used progerin-transfected HEK293 cells and demonstrated that LPA3 activation increases the expression of antioxidant enzymes such as SOD and GPx1, decreasing the damage produced by ROS. Additionally, these authors utilized siRNA to block LPA3 expression in mouse fibroblasts, and such treatment reduced the expression of these enzymes, increased ROS production, and induced cellular senescence [55].
These findings are particularly interesting because they suggest that LPA3 could be a target to treat diseases in which the participation of ROS promotes cell death. Evidence suggests that in Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, and other degenerative pathologies, increased ROS production is associated with cell death [56,57]. Studies employing a mouse Huntington’s model showed that the stimulation of LPA1 and LPA3 receptors by gintonin (a complex of LPA molecules and ginseng proteins such as Ginseng major latex-like protein 151) activates the Nrf2 pathway, which protects striatal neurons; the LPA1/3 inhibitor, Ki16425, blocked such protection [14].
However, the activation of the LPA3 receptors does not always induce a favorable outcome. It has been shown that the Nrf2 transcription factor can promote resistance to chemotherapy [58,59,60,61]. In HL60-DR cells, drug resistance was related to proliferation and a decrease in apoptosis; both events appearing to be related to the activation of the PI3K/AKT (protein kinase also named protein kinase B (PKB))/Nrf2 signal pathway [58,62]. It is interesting to mention that these authors propose a role for LPA3/Nrf2/antioxidant enzymes in these actions [62,63]. LPA3 receptors activated by their ligands might promote the activation of the Gαi/o protein, which activates PI3K, AKT, and Nrf2, translocation to the nucleus, regulating the transcription of antioxidant enzymes that might promote survival and proliferation.

2.2. LPA3 Actions in Cardiac Cells and Function

The double-blade (favorable/unfavorable) pattern of action of LPA3 has also been observed in the heart. It has been revealed that LPA3 activity participates in the survival of cardiomyocytes from neonatal rats during hypoxia by inhibiting the autophagy that occurs in this event [64,65]. In contrast, it was reported that LPA action induced cardiac dysfunction after myocardial infarction in a murine model. These processes could involve the regulation of the PI3K, Rho (Ras homologues, family of small GTPases), and AKT pathways, and the involvement of Gαi and Gα12/13 has been proposed [34,64,65,66,67].
Some studies have reported that the activation of LPA3 and β-adrenergic receptors promote cardiac hypertrophy; however, the signaling pathways involved appear to be different, at least in H9C2 cardiomyocytes [34]. When LPA3 expression is inhibited, an increase in the expression of atrial natriuretic peptide is produced, which inhibits cardiac dysfunction, after myocardial infarction [34]. Additional studies showed that LPA promotes cardiac hypertrophy in cardiomyocytes from neonatal rats and increases cellular apoptosis, cellular elongation, and actin fiber reorganization; it has been suggested that two signaling pathways are involved herein, one through PI3K/AKT/mTOR (protein kinase, mammalian Target Of Rapamycin)/ERK (Extracellular-Signal-Regulated Kinase) 1/2 and the other including NFκB [34,65,66].

2.3. Participation of LPA3 in Fertility, Embryo Implantation, and Development

During pregnancy, progesterone markedly increases in the blood, and many studies have shown that this hormone increases the expression of LPA3 mRNA. This LPA receptor participates in decidualization, implantation, oocyte maturation, and oviduct transport in mice and pigs [68,69]. LPA3 expression increases during the first stage of pregnancy, inducing decidualization related to the development of the placenta and the embryo. The importance of LPA3 has been evidenced by demonstrating that its knockdown decreases preimplantation with a loss of embryonic spacing, producing the implantation of 3–4 embryos in the same place. This rendered the reabsorption of many embryos, and those that managed to survive were small in size and weight [68,69,70,71]. In males, LPA3 and LPA2 are expressed in the seminiferous tubules, spermatogonia, and spermatocytes, and such expression is related to male fertility [70].

2.4. LPA3 in Cancer

Cancer is another pathology in which the expression and action of LPA3 receptors are usually associated with a poor prognosis. Studies on breast cancer have shown that LPA3 expression was related to metastasis and proliferation in cancer cells [27,72,73]. Several studies have reported that LPA3 is expressed in breast cancer tumors (grades 2 and 3), but the signaling pathway by which the receptor regulates this event is not yet fully known [27,72,74]. In addition, there is evidence that LPA3 receptors increase proliferation and migration in OVCAR-3 and SKOV3 cells and that these cells lose sensitivity to ultraviolet light and increase their survival rate [19,74,75,76].
Interestingly, LPA3 produces drug resistance in pancreatic and hepatic cancer cells. In pancreatic cancer, LPA3 expression is considered an indicator or marker of aggressiveness, including metastasis and accelerated regrowth [74,75]. In human sarcoma cells, and in pancreatic and hepatic cancer cells, LPA3 activation increases migration, invasion, and metastasis [28,77,78]. These processes are probably related to the increase in matrix metalloproteinases 9 expression, whose proteolytic activity produces the degradation of the extracellular matrix, favoring metastasis. In addition, in hepatoma cells, R777AB, LPA3 action is associated with the expression of genes related to drug resistance, such as Mdr (Multidrug resistance protein) 1a and 1b and Gstp1 (Glutathion-S-transferase pi 1) [15,77,78,79,80,81]. The signaling pathways that promote these processes appear to be related the action of Gαi/o and Gαq, because the inhibitors of these signaling pathways decrease the proliferation and expression of genes associated with drug resistance and the migration of these cancer cells [79].
It is noteworthy that the increased expression of the LPA3 receptors is related to a grim prognosis in cancer; however, this dark side might represent a portal to optimism, if the possibility of LPA3 becoming a target for therapeutic intervention is considered.

2.5. Other Processes in Which LPA3 Participates

LPA3 is a receptor that participates during neuritic ramifications after birth. This process is essential because it creates neural networks and maintains information fluxes within neuronal circuits. It has been reported that LPA3 activation increases neurite branching in the hippocampus and brain cortex of mice [82,83].
LPA3 appears to participate in the maturation of dendritic cells, which are specialized in the immune response. Antagonists and the genetic inhibition of LPA3 expression decrease chemotaxis in immature dendritic cells. The process could affect the maturation of these cells, compromising the immune response [32,84]. The mechanisms through which LPA3 regulates this process are currently unknown.
LPA3 activation also participates in inflammatory processes. LPA3 activation promotes the infiltration of interleukins such as IL-6, IL-16, and IL-8 and the synthesis of prostaglandin E2 in patients with rheumatoid arthritis and in fibroblast-like synoviocytes from patients with this disease [85,86]. The signaling pathway through which the receptor produced these events appears to involve Gαi/o, which activates the MAPK signaling cascade.

3. The LPA3 Receptor: Structure and Function

The human LPA3 receptor (https://www.uniprot.org/uniprot/Q9UBY5; Accessed on 12 May 2021) is constituted of 353 amino acids (mouse and rat orthologs, 354 amino acids), and its calculated molecular weight is ≈40 KDa (39,998 Da) [5,87,88]. As previously indicated, according to the classification systems GRAFS and A-F, this receptor belongs to the A family [37,38]. LPA3 is mainly coupled to two G proteins, Gαq/11 and Gαi/o; therefore, the G protein-binding motif of this receptor subtype is considered promiscuous. This property allows this receptor to activate different signal pathways, which might explain why it does participate in a large variety of physiological functions and, as previously mentioned, in the pathogenesis of diseases [5,8,89].
As a member of the GPCR superfamily, the LPA3 receptor is constituted of seven hydrophobic transmembrane domains (TM), which are joined together through three extracellular and three intracellular loops (Figure 5). It is worth mentioning that transmembrane regions are essential for this receptor, as has been observed for others that also belong to the A family. These regions or domains are frequently conserved [90]. For clarity, the sequence indicating the transmembrane domains is presented as (Supplementary Figure S1).
Available information on LPA3 receptor structure/function is scarce. Therefore, in order to obtain some information, we performed in silico analyses. This allowed us to identify different domains observed in other GPCRs. Among these are the following: an ERH (Glutamic acid-Arginine-Intrahelical hydrogen bonding residue) domain (analogous to the DRY (Aspartic acid-Arginine-Tyrosine) motif) in the transition between the end of TM3 and the initiation of ICL2, a CWXP domain within TM6, an NPXXY domain near the end of TM7, and a di-cysteine domain within the carboxyl terminus (Figure 5). Studies on these domains in other receptors have shown that they are important for the activation and regulation of the GPCRs receptors of the A family [91,92,93,94]. Additionally, an AP2-binding domain is present in the carboxyl terminus [94,95,96].
It is noteworthy to mention that the mutation of these domains usually reduces or abolishes agonist-activation of GPCRs. Studies employing molecular docking showed that ligand binding at GPCRs produced the packaging of TM3-5-6-7 domains; this event was promoted by destabilization of an ionic interaction [92,97], initiating a displacement of TM7 toward TM3 and promoting activation involving the tyrosine residue present in the DRY motif, which is associated with the rotation of the cytoplasmic extreme of TM6 and which promotes the activation of these receptors [92,98,99,100,101].
Additionally, the asparagine residue of the NPXXY motif establishes interactions with other residues, facilitating the movement of TM7 toward TM3 [92,99] and promoting the stability of the activated receptor. Finally, the DRY motif forms a salt bridge with surrounding residues and with TM6; this salt bridge breaks at the moment the ligand binds. The DRY motif creates a new interaction with TM5, stabilizing the receptor in its active conformation, breaking contacts between TM3 and TM6, thus promoting a movement toward the cellular cytoplasm of TM6, which increases the receptor binding to the Gα protein. These events initiate signaling, favor receptor phosphorylation, and later favor association with β-arrestins, all of which are relevant for receptor desensitization [53,92,99,100,101].
The CWXP domain is a motif found in TM6 which seems to participate in the binding of agonists. Rotation of the tryptophan residue causes movements within the binding pocket, promoting the accommodation of the ligand into the receptor. In contrast, the proline residue induces a bend that serves as a pivot for helical movement during receptor activation [92,93,99,101,102]. Other motifs that appear to participate in the activation of GPCRs include the PIF (GPCR microswitch; Proline-Isoleucine-Phenylalanine) motif that is usually found in TM4 and the NPXXY motif found in TM7, both of which are also related to the activation of Gαq, Gαs, Gαi and β-arrestins [99,103,104,105,106]. It has been shown that in some receptors (such as the histamine 2 receptor [106,107], the formyl peptide receptor [47,100,107], and α- and β-adrenoceptors [93,108], among others), this domain could be regulating agonist-induced internalization, which affects MAPK pathway activation and intracellular calcium mobilization.
The majority of the motifs that generally regulate the activation of GPCRs, including those in the LPA1 receptor, have also been found in the LPA3 receptor (Figure 5). Only the PIF domain could not be found in the receptor sequence. Therefore, it appears likely that other receptor region(s) could replace the role of PIF in receptor activation.
This illustrates the putative importance of the motifs present in the LPA3 receptor at the time of its activation when the ligand binds to it; however, we must recall that the intracellular loops and the carboxyl-terminal region play essential roles, particularly in receptor desensitization and internalization. Current ideas suggest key roles in the phosphorylation of specific residues, mediated by GRKs, second messenger-activated, and other protein kinases [100,109,110].
Other important regions of the LPA3 structure are the transmembrane domains, which contain residues that take part in ligand binding. It is worth mentioning that the LPA receptors that belong to the lysophospholipid subfamily entertain an ≈81% similarity among themselves [111,112].
Few studies have reported the participation of these residues during the binding of the ligand in LPA receptors. The residues where LPA has been shown to interact with LPA receptors include arginine 105, glutamine 106, tryptophan 153, arginine 185, lysine 279, and arginine 276 (Figure 5, residues in green). These sites are conserved in the LPA1, LPA2, and LPA3 receptors, but differences appear to exist between these [5,87,89,111,112]. In the case of tryptophan 153, when it was mutated to alanine in the LPA3 receptor, it induced a decrease in the potency and efficacy of LPA; such changes were not observed when the LPA1 and LPA2 receptors were similarly mutated. Likewise, when arginine 279 was substituted with alanine, a decrease in the activation of LPA1 and LPA2, but not in the LPA3 receptor, was observed [111,112].
An amphipathic α-helix is found in the carboxyl terminus of many of the GPCRs of the A family. It is frequently denominated helix 8, and it has a conserved sequence in nearly all of these receptors, i.e., F (R/K) XX (F/L) XXX (L/F); it has been shown that it allows maintaining the surface expression of these GPCRs, promoting GPCR trafficking, and participating in the activation of the G proteins and the receptor’s interaction with the β-arrestins [113,114,115,116].
Zhou and coworkers suggested a mechanism through which many receptors belonging to the GPCR A family could recruit G proteins. It was proposed that in response to agonist-induced conformational changes, residues in transmembrane domains 3, 5, and 6 interact with and activate G proteins [109]. These residues were found in the structure of the LPA3 receptor as shown in Figure 5 (indicated in cerulean).
The GRKs are a family of protein kinases that appears to play a major role in the phosphorylation of agonist-occupied GPCRs (Table 1). This family is made up of seven different isoforms that are constituted of a central catalytic domain which is conserved in all GRKs; an amino-terminal area and the carboxyl terminus, both of which differ among these protein kinases, seem to confer them selectivity in their action, and participate in their regulation. These domains constitute the structural basis for their classification into subfamilies; in addition, some GRKs exhibit selective expression in some tissues [117,118,119,120]. The visual GRKs (GRK1 and GRK7) are mainly expressed in the retina, GRK4 is mainly expressed in the testis, whereas the other GRKs (2, 3, 5, and 6) are ubiquitously expressed; visual GRKs have short prenylation sequences (see reviews in [117,120] and references therein). The second subfamily, denominated GRK2 and also, for historical reasons, the β-adrenergic receptor kinase (or βARK) subfamily, exhibits a Pleckstrin homology domain that interacts with G protein βγ dimers and phosphatidylinositol 4, 5-bisphosphate. These kinases are cytoplasmic and their interaction with the plasma membrane seems to occur through these domains. The GRK4 subfamily seems to be bound to the plasma membrane through palmitoylation and/or the presence of positively charged lipid binding elements [117,118,119,120]. It has been proposed that lipids covalently bound to the carboxyl terminus of these proteins, the Pleckstrin homology domain that associates with phosphoinositides, and the polybasic/hydrophobic regions permit these kinases to be recruited to the membrane and to catalyze GPCR phosphorylation at specific residues [119,120,121,122,123].
Such specificity in the GPCR phosphorylation pattern appears to be critical to define subsequent signaling (frequently associated with β-arrestin activation), vesicular trafficking, and the receptor’s fate (rapid or slow recycling to the plasma membrane, or degradation). This has been named the “GPCR phosphorylation barcode,” and numerous research groups are actively working to understand (i.e., to break) this code, which currently is only partially understood [46,50,124,125,126,127,128]. Obviously, initial steps include knowing that the GPCR of interest is actually phosphorylated, the conditions under which that takes place, and the definition of the specific sites affected by such covalent modification. At present, there is evidence that LPA3 receptors are phosphorylated in response to agonists and other agents (associated respectively with homologous and heterologous desensitizations) [46,89]. However, to date, the phosphorylation pattern(s) of this receptor is (are) unknown, which seems to be an important gap in our knowledge.
Studies conducted in silico showed that the LPA3 receptor can be phosphorylated by different protein kinases [89]. Not surprisingly, different isoforms of GRK and PKC are predicted to be responsible for many such phosphorylations; however, other protein kinases such as PKA, PKB/AKT, and some protein tyrosine kinases were present in this in silico analysis [129]. Many of these predicted phosphorylation sites could be targeted by several protein kinases [89,129].
Considering the vital role that GRKs play in homologous desensitization/phosphorylation, the putative sites for the action of this family of kinases on LPA3 receptor phosphorylation are presented in Figure 6. These residues were obtained in a new analysis employing different and/or updated software programs, including GPS5 (http://gps.biocuckoo.cn; Accessed on 3 April 2021), netphorest (http://netphorest.info; Accessed on 3 April 2021), quokka (https://quokka.erc; Accessed on 4 April 2021) and NetPhos 3.1 (http://www.cbs.dtu.dk; Accessed on 4 April 2021). The criterion used to carry out each study was a high threshold. Only residues that were putative targets of GRK, PKA, or PKC and that obtained a high score were considered. Subsequently, we carried out an analysis on the results obtained and chose the residues that were consistently observed in these analyses; these are presented in Figure 6. The majority of the GRK putative phosphorylation-target residues were found in intracellular loop 3 and the carboxyl terminus region. Not surprisingly, the different software programs used suggested roles of isoforms of the GRK2 and GRK4 subfamilies (Table 1).
The possibility that different GRK isoforms may participate in LPA3 phosphorylation is provocative. It has been proposed that GRK 2 and 3 promote receptor endocytosis by the β-arrestin/clathrin pathway more efficiently than other isoforms. At the same time, GRK 5 and 6 appear to mediate β-arrestin-triggered ERK 1/2 signaling [78,117,119,125,130,131,132,133,134]. It is important to mention that GRKs, in addition to carrying out GPCR phosphorylation, can phosphorylate other proteins in the cell cytoplasm that are involved in cell signaling, as well as receptor trafficking proteins such as Gαq and Gβγ, PI3K, clathrin, caveolin, MEK, and AKT/PKB, among others [123,124,135,136,137,138,139,140].
It is noteworthy that the in silico analysis suggested that PKA and PKC could participate in LPA3 receptor phosphorylation (Figure 6 and Table 2); this result is of interest because it might indicate the involvement of these protein kinases in the heterologous desensitization of this receptor. It has been reported previously that LPA1–3 receptors can be phosphorylated in response to the pharmacological activation of PCK with phorbol myristate acetate [89]. However, to the extent of our knowledge, there is no evidence of PKA-induced LPA3 receptor phosphorylation. It should be noted that the in silico analysis revealed marked overlapping between GRKs, PKA and PKC, suggesting that some sites could be targeted by these groups of kinases (Table 1 and Table 2).

4. The Regulation of LPA3 Receptors and Its Possible Impact on Signaling

It is worth considering that LPA3 receptor phosphorylation has not, to our knowledge, been studied in detail and that the sites actually phosphorylated in whole cells under different stimuli are presently unknown. Similarly, the LPA3 receptor internalization process is essentially unexplored. Therefore, we have to assume that the molecular consequences of phosphorylation in signaling and intracellular location/trafficking are similar to those observed for other receptors. Obviously, such assumptions are plainly hypothetical and require experimental confirmation. The in silico analysis of the LPA3 receptor structure indicates many putative phosphorylation sites, mainly located in intracellular loop 3 and the carboxyl terminus. It seems, therefore, reasonable to suggest that several patterns of phosphorylation might exist and that these might affect signaling, action outcomes, and vesicular traffic. Thus, their study might contribute to a better understanding of the GPCR phosphorylation bar code [89,129].
It has been shown that agonist activation promotes conformational movement of GPCRs, exposing sites that are phosphorylated by GRKs and revealing that such phosphorylations favor the recruitment of β-arrestins [109,110,122,141]. This process has been associated with the phosphorylation patterns mainly observed at the carboxyl termini of GPCRs of the A and B families. In particular, the TXXS motif appears to be of importance for receptor binding to β-arrestins [110,142,143]. Our in silico analysis of the LPA3 receptor indicates that a domain of this type is present between threonine 326 and serine 329 (TVLS) at the carboxyl terminus (Figure 6); threonine 326 is a putative phosphorylation site of GRKs (Figure 6). It has been shown that the mutation of equivalent sites decreases the interaction of β-arrestins with other GPCRs [109,110,142].
GPCR binding with β-arrestins promotes the recruitment of proteins such as clathrin, dynamin, Adaptin 2, Eps 15, and Rab5, which belong to the endocytic machinery, promoting receptor internalization and early endosome formation. This is followed by internalization with the clathrin assembly lymphoid myeloid leukemia (CALM) protein, as shown in the Epidermal Growth Factor receptor, the LPA1 receptor, and other receptors [51,144].
It is noteworthy that Urs and coworkers [143] demonstrated that the LPA1 receptor is internalized through different mechanisms when agonist-activated as compared to when it is in response to PKC pharmacological activation; a dileucine domain is critical for PKC-activation induced receptor internalization. No dileucine motif was detected in the LPA3 structure. However, other sites appear to be related to internalization, including the following: YXXφ (φ, hydrophobic amino acid; X, any other amino acid); YXT; YXXL; DEXXXLI or DXXLL; NPXY; GDAY; and DIEXXXLL [145,146,147,148,149,150,151]. The YXT domain and the NPXY domain are present in the LPA3 receptor. These motifs might participate in LPA3 receptor endocytosis, but this must be verified experimentally.
It has been shown that PDZ (structural domain of 80–90 amino-acids; Post-synaptic density protein 95 (PSD-95), Drosophila disc large tumor suppressor (Dlg1), and Zona occludens 1 (ZO-1)) domains participate in the regulation of GPCRs [144,145,146,147,148,149,150,151,152]. In the case of the LPA1 receptor, the PDZ domain binds to the GIPC protein, which regulates the function of the receptor and its trafficking, in that both decreasing the expression of GIPC and mutating the PDZ domain induced deficient receptor internalization, promoting its constant activation, and increasing cell proliferation and migration [153]. This event could be directly related to the metastatic processes with which LPA1 is associated. Likewise, LPA2 has been shown to present a PDZ domain to which the NHERF-2 and MAGI-3 proteins bind, and that could be involved in tumorigenesis, cell invasion, migration, and inflammation [154,155]. The LPA3 receptor does not appear to present any canonical PDZ domain.
At present, much is unknown about the regulation of LPA3 receptors, which, although part of the LPA receptor family, structurally present possible differential regulatory sites with the other receptors. We conclude that LPA3 remains an enigmatic receptor, about which we are just beginning to learn its functions in health and roles in the pathogenesis of diseases but have very little knowledge on its regulation.

Supplementary Materials

Author Contributions

K.H.S. originated the review and wrote the initial draft. The other authors (M.T.R.-Á., A.G.-S. and J.A.G.-S.) participated by adding comments and suggestions and corrected the initial draft and its organization. All authors have read and agreed to the published version of the manuscript.

Funding

Work on LPA receptors in our laboratory is partially supported by Grants from CONACyT (Fronteras 6676) and DGPA (IN201221). K.H. Solís is a Ph.D. student in the Programa de Doctorado en Ciencias Biomédicas and is supported by a fellowship from CONACyT CVU (508696). A. Guzmán-Silva is a Ph.D. student in the Programa de Doctorado en Ciencias Bioquímicas and is supported by a fellowship from CONACyT CVU (380520).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors express their gratitude to Maggie Brunner, M.A., for editorial/style corrections.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kihara, Y.; Maceyka, M.; Spiegel, S.; Chun, J. Lysophospholipid Receptor Nomenclature Review: IUPHAR Review 8: Lysophospholipid Receptor Nomenclature. Br. J. Pharmacol. 2014, 171, 3575–3594. [Google Scholar] [CrossRef] [Green Version]
  2. Yang, F.; Chen, G.-X. Production of Extracellular Lysophosphatidic Acid in the Regulation of Adipocyte Functions and Liver Fibrosis. World J. Gastroenterol. 2018, 24, 4132–4151. [Google Scholar] [CrossRef] [PubMed]
  3. Gaits, F.; Fourcade, O.; Le Balle, F.; Gueguen, G.; Gaigé, B.; Gassama-Diagne, A.; Fauvel, J.; Salles, J.P.; Mauco, G.; Simon, M.F.; et al. Lysophosphatidic Acid as a Phospholipid Mediator: Pathways of Synthesis. FEBS Lett. 1997, 410, 54–58. [Google Scholar] [CrossRef] [Green Version]
  4. Nakanaga, K.; Hama, K.; Aoki, J. Autotaxin-An LPA Producing Enzyme with Diverse Functions. J. Biochem. 2010, 148, 13–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Yung, Y.C.; Stoddard, N.C.; Chun, J. LPA Receptor Signaling: Pharmacology, Physiology, and Pathophysiology. J. Lipid Res. 2014, 55, 1192–1214. [Google Scholar] [CrossRef] [Green Version]
  6. Aoki, J. Mechanisms of Lysophosphatidic Acid Production. Semin. Cell Dev. Biol. 2004, 15, 477–489. [Google Scholar] [CrossRef] [PubMed]
  7. Ramesh, S.; Govindarajulu, M.; Suppiramaniam, V.; Moore, T.; Dhanasekaran, M. Autotaxin–Lysophosphatidic Acid Signaling in Alzheimer’s Disease. Int. J. Mol. Sci. 2018, 19, 1827. [Google Scholar] [CrossRef] [Green Version]
  8. Choi, J.W.; Herr, D.R.; Noguchi, K.; Yung, Y.C.; Lee, C.-W.; Mutoh, T.; Lin, M.-E.; Teo, S.T.; Park, K.E.; Mosley, A.N.; et al. LPA Receptors: Subtypes and Biological Actions. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 157–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Chun, J.; Hla, T.; Lynch, K.R.; Spiegel, S.; Moolenaar, W.H. International Union of Basic and Clinical Pharmacology. LXXVIII. Lysophospholipid Receptor Nomenclature: TABLE 1. Pharmacol. Rev. 2010, 62, 579–587. [Google Scholar] [CrossRef] [Green Version]
  10. Yasuda, D.; Kobayashi, D.; Akahoshi, N.; Ohto-Nakanishi, T.; Yoshioka, K.; Takuwa, Y.; Mizuno, S.; Takahashi, S.; Ishii, S. Lysophosphatidic Acid–Induced YAP/TAZ Activation Promotes Developmental Angiogenesis by Repressing Notch Ligand Dll4. J. Clin. Investig. 2019, 129, 4332–4349. [Google Scholar] [CrossRef]
  11. Rivera-Lopez, C.M.; Tucker, A.L.; Lynch, K.R. Lysophosphatidic Acid (LPA) and Angiogenesis. Angiogenesis 2008, 11, 301–310. [Google Scholar] [CrossRef] [Green Version]
  12. Gross, I.; Bräuer, A. Modulation of Lysophosphatidic Acid (LPA) Receptor Activity: The Key to Successful Neural Regeneration? Neural Regen. Res. 2020, 15, 53. [Google Scholar] [CrossRef]
  13. Sayas, C.L.; Moreno-Flores, M.T.; Avila, J.; Wandosell, F. The Neurite Retraction Induced by Lysophosphatidic Acid Increases Alzheimer’s Disease-like Tau Phosphorylation. J. Biol. Chem. 1999, 274, 37046–37052. [Google Scholar] [CrossRef] [Green Version]
  14. Jang, M.; Choi, J.H.; Chang, Y.; Lee, S.J.; Nah, S.-Y.; Cho, I.-H. Gintonin, a Ginseng-Derived Ingredient, as a Novel Therapeutic Strategy for Huntington’s Disease: Activation of the Nrf2 Pathway through Lysophosphatidic Acid Receptors. Brain Behav. Immun. 2019, 80, 146–162. [Google Scholar] [CrossRef] [PubMed]
  15. Tanabe, E.; Kitayoshi, M.; Yoshikawa, K.; Shibata, A.; Honoki, K.; Fukushima, N.; Tsujiuchi, T. Loss of Lysophosphatidic Acid Receptor-3 Suppresses Cell Migration Activity of Human Sarcoma Cells. J. Recept. Signal. Transduct. 2012, 32, 328–334. [Google Scholar] [CrossRef] [PubMed]
  16. Okabe, K.; Kato, K.; Teranishi, M.; Okumura, M.; Fukui, R.; Mori, T.; Fukushima, N.; Tsujiuchi, T. Induction of Lysophosphatidic Acid Receptor-3 by 12-O-Tetradecanoylphorbol-13-Acetate Stimulates Cell Migration of Rat Liver Cells. Cancer Lett. 2011, 309, 236–242. [Google Scholar] [CrossRef]
  17. Shano, S.; Hatanaka, K.; Ninose, S.; Moriyama, R.; Tsujiuchi, T.; Fukushima, N. A Lysophosphatidic Acid Receptor Lacking the PDZ-Binding Domain Is Constitutively Active and Stimulates Cell Proliferation. Biochim. Biophys. Acta BBA Mol. Cell Res. 2008, 1783, 748–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Hayashi, M.; Okabe, K.; Kato, K.; Okumura, M.; Fukui, R.; Fukushima, N.; Tsujiuchi, T. Differential Function of Lysophosphatidic Acid Receptors in Cell Proliferation and Migration of Neuroblastoma Cells. Cancer Lett. 2012, 316, 91–96. [Google Scholar] [CrossRef]
  19. Goldsmith, Z.G.; Ha, J.H.; Jayaraman, M.; Dhanasekaran, D.N. Lysophosphatidic Acid Stimulates the Proliferation of Ovarian Cancer Cells via the Gep Proto-Oncogene G 12. Genes Cancer 2011, 2, 563–575. [Google Scholar] [CrossRef] [Green Version]
  20. Anliker, B.; Choi, J.W.; Lin, M.-E.; Gardell, S.E.; Rivera, R.R.; Kennedy, G.; Chun, J. Lysophosphatidic Acid (LPA) and Its Receptor, LPA1, Influence Embryonic Schwann Cell Migration, Myelination, and Cell-to-Axon Segregation: LPA 1 Regulates Schwann Cell Physiology. Glia 2013, 61, 2009–2022. [Google Scholar] [CrossRef] [Green Version]
  21. Sánchez-Marín, L.; Ladrón de Guevara-Miranda, D.; Mañas-Padilla, M.C.; Alén, F.; Moreno-Fernández, R.D.; Díaz-Navarro, C.; Pérez-del Palacio, J.; García-Fernández, M.; Pedraza, C.; Pavón, F.J.; et al. Systemic Blockade of LPA1/3 Lysophosphatidic Acid Receptors by ki16425 Modulates the Effects of Ethanol on the Brain and Behavior. Neuropharmacology 2018, 133, 189–201. [Google Scholar] [CrossRef]
  22. Yung, Y.C.; Stoddard, N.C.; Mirendil, H.; Chun, J. Lysophosphatidic Acid Signaling in the Nervous System. Neuron 2015, 85, 669–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ueda, H.; Neyama, H.; Sasaki, K.; Miyama, C.; Iwamoto, R. Lysophosphatidic Acid LPA1 and LPA3 Receptors Play Roles in the Maintenance of Late Tissue Plasminogen Activator-Induced Central Poststroke Pain in Mice. Neurobiol. Pain 2019, 5, 100020. [Google Scholar] [CrossRef]
  24. Kuwajima, K.; Sumitani, M.; Kurano, M.; Kano, K.; Nishikawa, M.; Uranbileg, B.; Tsuchida, R.; Ogata, T.; Aoki, J.; Yatomi, Y.; et al. Lysophosphatidic Acid Is Associated with Neuropathic Pain Intensity in Humans: An Exploratory Study. PLoS ONE 2018, 13, e0207310. [Google Scholar] [CrossRef] [PubMed]
  25. Fayyaz, S.; Japtok, L.; Schumacher, F.; Wigger, D.; Schulz, T.J.; Haubold, K.; Gulbins, E.; Völler, H.; Kleuser, B. Lysophosphatidic Acid Inhibits Insulin Signaling in Primary Rat Hepatocytes via the LPA3 Receptor Subtype and Is Increased in Obesity. Cell. Physiol. Biochem. 2017, 43, 445–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Tigyi, G.; Dacheux, M.A.; Lin, K.H.; Yue, J.; Norman, D.; Benyó, Z.; Lee, S.C. Anti-Cancer Strategies Targeting the Autotaxin-Lysophosphatidic Acid Receptor Axis: Is There a Path Forward? Cancer Metastasis Rev. 2021, 40, 3–5. [Google Scholar] [CrossRef] [PubMed]
  27. Sun, K.; Cai, H.; Duan, X.; Yang, Y.; Li, M.; Qu, J.; Zhang, X.; Wang, J. Aberrant Expression and Potential Therapeutic Target of Lysophosphatidic Acid Receptor 3 in Triple-Negative Breast Cancers. Clin. Exp. Med. 2015, 15, 371–380. [Google Scholar] [CrossRef] [Green Version]
  28. Quan, M.; Cui, J.; Feng, X.; Huang, Q. The Critical Role and Potential Target of the Autotaxin/Lysophosphatidate Axis in Pancreatic Cancer. Tumor Biol. 2017, 39, 101042831769454. [Google Scholar] [CrossRef] [Green Version]
  29. Chen, J.; Li, H.; Xu, W.; Guo, X. Evaluation of Serum ATX and LPA as Potential Diagnostic Biomarkers in Patients with Pancreatic Cancer. BMC Gastroenterol. 2021, 21, 58. [Google Scholar] [CrossRef]
  30. Lee, J.; Kim, D.; Oh, Y.; Jun, H.-S. Lysophosphatidic Acid Signaling in Diabetic Nephropathy. Int. J. Mol. Sci. 2019, 20, 2850. [Google Scholar] [CrossRef] [Green Version]
  31. Tigyi, G.; Miledi, R. Lysophosphatidates Bound to Serum Albumin Activate Membrane Currents in Xenopus Oocytes and Neurite Retraction in PC12 Pheochromocytoma Cells. J. Biol. Chem. 1992, 267, 21360–21367. [Google Scholar] [CrossRef]
  32. Panther, E.; Idzko, M.; Corinti, S.; Ferrari, D.; Herouy, Y.; Mockenhaupt, M.; Dichmann, S.; Gebicke-Haerter, P.; di Virgilio, F.; Girolomoni, G.; et al. The Influence of Lysophosphatidic Acid on the Functions of Human Dendritic Cells. J. Immunol. 2002, 169, 4129–4135. [Google Scholar] [CrossRef] [Green Version]
  33. Ward, J.D.; Dhanasekaran, D.N. LPA Stimulates the Phosphorylation of P130Cas via GI2 in Ovarian Cancer Cells. Genes Cancer 2012, 3, 578–591. [Google Scholar] [CrossRef] [PubMed]
  34. Yang, J.; Xu, J.; Han, X.; Wang, H.; Zhang, Y.; Dong, J.; Deng, Y.; Wang, J. Lysophosphatidic Acid Is Associated with Cardiac Dysfunction and Hypertrophy by Suppressing Autophagy via the LPA3/AKT/MTOR Pathway. Front. Physiol. 2018, 9, 1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Liao, Y.; Mu, G.; Zhang, L.; Zhou, W.; Zhang, J.; Yu, H. Lysophosphatidic Acid Stimulates Activation of Focal Adhesion Kinase and Paxillin and Promotes Cell Motility, via LPA1–3, in Human Pancreatic Cancer. Dig. Dis. Sci. 2013, 58, 3524–3533. [Google Scholar] [CrossRef]
  36. Aoki, J. Two Pathways for Lysophosphatidic Acid Production. Biochim. Biophys. Acta (BBA) 2008, 1781, 513–518. [Google Scholar] [CrossRef]
  37. Schiöth, H.B.; Fredriksson, R. The GRAFS Classification System of G-Protein Coupled Receptors in Comparative Perspective. Gen. Comp. Endocrinol. 2005, 142, 94–101. [Google Scholar] [CrossRef] [PubMed]
  38. Davies, M.N.; Secker, A.; Freitas, A.A.; Mendao, M.; Timmis, J.; Flower, D.R. On the Hierarchical Classification of G Protein-Coupled Receptors. Bioinformatics 2007, 23, 3113–3118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Takuwa, Y.; Takuwa, N.; Sugimoto, N. The Edg Family G Protein-Coupled Receptors for Lysophospholipids: Their Signaling Properties and Biological Activities. J. Biochem. 2002, 131, 767–771. [Google Scholar] [CrossRef] [PubMed]
  40. Neve, K.A.; Seamans, J.K.; Trantham-Davidson, H. Dopamine Receptor Signaling. J. Recept. Signal. Transduct. 2004, 24, 165–205. [Google Scholar] [CrossRef] [PubMed]
  41. Lin, M.-E.; Herr, D.R.; Chun, J. Lysophosphatidic Acid (LPA) Receptors: Signaling Properties and Disease Relevance. Prostaglandins Other Lipid Mediat. 2010, 91, 130–138. [Google Scholar] [CrossRef] [Green Version]
  42. Dalle, S.; Imamura, T.; Rose, D.W.; Worrall, D.S.; Ugi, S.; Hupfeld, C.J.; Olefsky, J.M. Insulin Induces Heterologous Desensitization of G Protein-Coupled Receptor and Insulin-Like Growth Factor I Signaling by Downregulating β-Arrestin-1. Mol. Cell. Biol. 2002, 22, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Calebiro, D.; Godbole, A. Internalization of G-Protein-Coupled Receptors: Implication in Receptor Function, Physiology and Diseases. Best Pract. Res. Clin. Endocrinol. Metab. 2018, 32, 83–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Liu, Q.; Bee, M.S.; Schonbrunn, A. Site Specificity of Agonist and Second Messenger-Activated Kinases for Somatostatin Receptor Subtype 2A (Sst2A) Phosphorylation. Mol. Pharmacol. 2009, 76, 68–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Premont, R.T.; Inglese, J.; Lefkowitz, R.J. Protein Kinases That Phosphorylate Activated G Protein-coupled Receptors. FASEB J. 1995, 9, 175–182. [Google Scholar] [CrossRef] [PubMed]
  46. Alfonzo-Méndez, M.A.; Carmona-Rosas, G.; Hernández-Espinosa, D.A.; Romero-Ávila, M.T.; García-Sáinz, J.A. Different Phosphorylation Patterns Regulate A1D-Adrenoceptor Signaling and Desensitization. Biochim. Biophys. Acta (BBA) 2018, 1865, 842–854. [Google Scholar] [CrossRef]
  47. Wootten, D.; Christopoulos, A.; Marti-Solano, M.; Babu, M.M.; Sexton, P.M. Mechanisms of Signalling and Biased Agonism in G Protein-Coupled Receptors. Nat. Rev. Mol. Cell Biol. 2018, 19, 638–653. [Google Scholar] [CrossRef]
  48. Mundell, S.J.; Jones, M.L.; Hardy, A.R.; Barton, J.F.; Beaucourt, S.M.; Conley, P.B.; Poole, A.W. Distinct Roles for Protein Kinase C Isoforms in Regulating Platelet Purinergic Receptor Function. Mol. Pharmacol. 2006, 70, 1132–1142. [Google Scholar] [CrossRef] [Green Version]
  49. Murph, M.M.; Scaccia, L.A.; Volpicelli, L.A.; Radhakrishna, H. Agonist-Induced Endocytosis of Lysophosphatidic Acid-Coupled LPA1/EDG-2 Receptors via a Dynamin2- and Rab5-Dependent Pathway. J. Cell Sci. 2003, 116, 1969–1980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Magalhaes, A.C.; Dunn, H.; Ferguson, S.S. Regulation of GPCR Activity, Trafficking and Localization by GPCR-Interacting Proteins: Regulation of G-Protein-Coupled Receptor Activity. Br. J. Pharmacol. 2012, 165, 1717–1736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Huang, F.; Khvorova, A.; Marshall, W.; Sorkin, A. Analysis of Clathrin-Mediated Endocytosis of Epidermal Growth Factor Receptor by RNA Interference. J. Biol. Chem. 2004, 279, 16657–16661. [Google Scholar] [CrossRef] [Green Version]
  52. Lobingier, B.T.; von Zastrow, M. When Trafficking and Signaling Mix: How Subcellular Location Shapes G Protein-Coupled Receptor Activation of Heterotrimeric G Proteins. Traffic 2019, 20, 130–136. [Google Scholar] [CrossRef] [Green Version]
  53. Böhm, S.K.; Khitin, L.M.; Smeekens, S.P.; Grady, E.F.; Payan, D.G.; Bunnett, N.W. Identification of Potential Tyrosine-Containing Endocytic Motifs in the Carboxyl-Tail and Seventh Transmembrane Domain of the Neurokinin 1 Receptor. J. Biol. Chem. 1997, 272, 2363–2372. [Google Scholar] [CrossRef] [Green Version]
  54. Zaslavsky, A.; Singh, L.S.; Tan, H.; Ding, H.; Liang, Z.; Xu, Y. Homo- and Hetero-Dimerization of LPA/S1P Receptors, OGR1 and GPR4. Biochim. Biophys. Acta (BBA) 2006, 1761, 1200–1212. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, W.M.; Chiang, J.C.; Lin, Y.C.; Lin, Y.N.; Chuang, P.Y.; Chang, Y.C.; Chen, C.C.; Wu, K.Y.; Hsieh, J.C.; Chen, S.K.; et al. Lysophosphatidic Acid Receptor LPA3 Prevents Oxidative Stress and Cellular Senescence in Hutchinson–Gilford Progeria Syndrome. Aging Cell 2020, 19, 1–15. [Google Scholar] [CrossRef] [Green Version]
  56. Umeno, A.; Biju, V.; Yoshida, Y. In Vivo ROS Production and Use of Oxidative Stress-Derived Biomarkers to Detect the Onset of Diseases Such as Alzheimer’s Disease, Parkinson’s Disease, and Diabetes. Free Radic. Res. 2017, 51, 413–427. [Google Scholar] [CrossRef]
  57. Kolodkin, A.N.; Sharma, R.P.; Colangelo, A.M.; Ignatenko, A.; Martorana, F.; Jennen, D.; Briedé, J.J.; Brady, N.; Barberis, M.; Mondeel, T.D.G.A.; et al. ROS Networks: Designs, Aging, Parkinson’s Disease and Precision Therapies. NPJ Syst. Biol. Appl. 2020, 6, 34. [Google Scholar] [CrossRef] [PubMed]
  58. Bobilev, I.; Novik, V.; Levi, I.; Shpilberg, O.; Levy, J.; Sharoni, Y.; Studzinski, G.P.; Danilenko, M. The Nrf2 Transcription Factor Is a Positive Regulator of Myeloid Differentiation of Acute Myeloid Leukemia Cells. Cancer Biol. Ther. 2011, 11, 317–329. [Google Scholar] [CrossRef]
  59. Mirzaei, S.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Saleki, H.; Azami, N.; Hamzehlou, S.; Farahani, M.V.; Hushmandi, K.; Ashrafizadeh, M.; et al. Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery. Antioxidants 2021, 10, 349. [Google Scholar] [CrossRef]
  60. Wang, X.-J.; Sun, Z.; Villeneuve, N.F.; Zhang, S.; Zhao, F.; Li, Y.; Chen, W.; Yi, X.; Zheng, W.; Wondrak, G.T.; et al. Nrf2 Enhances Resistance of Cancer Cells to Chemotherapeutic Drugs, the Dark Side of Nrf2. Carcinogenesis 2008, 29, 1235–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Sun, Z.; Huang, G.; Cheng, H. Transcription Factor Nrf2 Induces the Up-Regulation of LncRNA TUG1 to Promote Progression and Adriamycin Resistance in Urothelial Carcinoma of the Bladder. Cancer Manag. Res. 2019, 11, 6079–6090. [Google Scholar] [CrossRef] [Green Version]
  62. Li, Y.; Guo, Y.; Feng, Z.; Bergan, R.; Li, B.; Qin, Y.; Zhao, L.; Zhang, Z.; Shi, M. Involvement of the PI3K/Akt/Nrf2 Signaling Pathway in Resveratrol-Mediated Reversal of Drug Resistance in HL-60/ADR Cells. Nutr. Cancer 2019, 71, 1007–1018. [Google Scholar] [CrossRef]
  63. Wang, L.; Chen, Y.; Sternberg, P.; Cai, J. Essential Roles of the PI3 Kinase/Akt Pathway in Regulating Nrf2-Dependent Antioxidant Functions in the RPE. Investig. Opthalmol. Vis. Sci. 2008, 49, 1671. [Google Scholar] [CrossRef] [Green Version]
  64. Hilal-Dandan, R.; Means, C.K.; Gustafsson, Å.B.; Morissette, M.R.; Adams, J.W.; Brunton, L.L.; Heller Brown, J. Lysophosphatidic Acid Induces Hypertrophy of Neonatal Cardiac Myocytes via Activation of Gi and Rho. J. Mol. Cell. Cardiol. 2004, 36, 481–493. [Google Scholar] [CrossRef]
  65. Chen, J.; Chen, Y.; Zhu, W.; Han, Y.; Han, B.; Xu, R.; Deng, L.; Cai, Y.; Cong, X.; Yang, Y.; et al. Specific LPA Receptor Subtype Mediation of LPA-Induced Hypertrophy of Cardiac Myocytes and Involvement of Akt and NFκB Signal Pathways. J. Cell. Biochem. 2008, 103, 1718–1731. [Google Scholar] [CrossRef]
  66. Cai, L.; Fan, G.; Wang, F.; Liu, S.; Li, T.; Cong, X.; Chun, J.; Chen, X. Protective Role for LPA3 in Cardiac Hypertrophy Induced by Myocardial Infarction but Not by Isoproterenol. Front. Physiol. 2017, 8, 356. [Google Scholar] [CrossRef] [PubMed]
  67. Kano, K.; Matsumoto, H.; Inoue, A.; Yukiura, H.; Kanai, M.; Chun, J.; Ishii, S.; Shimizu, T.; Aoki, J. Molecular Mechanism of Lysophosphatidic Acid-Induced Hypertensive Response. Sci. Rep. 2019, 9, 2662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Aikawa, S.; Kano, K.; Inoue, A.; Wang, J.; Saigusa, D.; Nagamatsu, T.; Hirota, Y.; Fujii, T.; Tsuchiya, S.; Taketomi, Y.; et al. Autotaxin–Lysophosphatidic Acid–LPA3 Signaling at the Embryo-epithelial Boundary Controls Decidualization Pathways. EMBO J. 2017, 36, 2146–2160. [Google Scholar] [CrossRef] [PubMed]
  69. Jeong, W.; Seo, H.; Sung, Y.; Ka, H.; Song, G.; Kim, J. Lysophosphatidic Acid (LPA) Receptor 3-Mediated LPA Signal Transduction Pathways: A Possible Relationship with Early Development of Peri-Implantation Porcine Conceptus. Biol. Reprod. 2016, 94. [Google Scholar] [CrossRef] [PubMed]
  70. Ye, X.; Hama, K.; Contos, J.J.A.; Anliker, B.; Inoue, A.; Skinner, K.; Suzuki, H.; Amano, T.; Kennedy, G.; Arai, H.; et al. LPA3-Mediated Lysophosphatidic Acid Signalling in Implantation and Embryo Spacing. Nature 2005, 435, 104–108. [Google Scholar] [CrossRef]
  71. Hama, K.; Aoki, J.; Bandoh, K.; Inoue, A.; Endo, T.; Amano, T.; Suzuki, H.; Arai, H. Lysophosphatidic Receptor, LPA3, Is Positively and Negatively Regulated by Progesterone and Estrogen in the Mouse Uterus. Life Sci. 2006, 79, 1736–1740. [Google Scholar] [CrossRef]
  72. Zhang, H.; Xu, X.; Gajewiak, J.; Tsukahara, R.; Fujiwara, Y.; Liu, J.; Fells, J.I.; Perygin, D.; Parrill, A.L.; Tigyi, G.; et al. Dual Activity Lysophosphatidic Acid Receptor Pan-Antagonist/Autotaxin Inhibitor Reduces Breast Cancer Cell Migration In Vitro and Causes Tumor Regression In Vivo. Cancer Res. 2009, 69, 5441–5449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Popnikolov, N.K.; Dalwadi, B.H.; Thomas, J.D.; Johannes, G.J.; Imagawa, W.T. Association of Autotaxin and Lysophosphatidic Acid Receptor 3 with Aggressiveness of Human Breast Carcinoma. Tumor Biol. 2012, 33, 2237–2243. [Google Scholar] [CrossRef] [PubMed]
  74. Yu, S.; Murph, M.M.; Lu, Y.; Liu, S.; Hall, H.S.; Liu, J.; Stephens, C.; Fang, X.; Mills, G.B. Lysophosphatidic Acid Receptors Determine Tumorigenicity and Aggressiveness of Ovarian Cancer Cells. JNCI J. Natl. Cancer Inst. 2008, 100, 1630–1642. [Google Scholar] [CrossRef] [Green Version]
  75. Fukushima, K.; Takahashi, K.; Yamasaki, E.; Onishi, Y.; Fukushima, N.; Honoki, K.; Tsujiuchi, T. Lysophosphatidic Acid Signaling via LPA1 and LPA3 Regulates Cellular Functions during Tumor Progression in Pancreatic Cancer Cells. Exp. Cell Res. 2017, 352, 139–145. [Google Scholar] [CrossRef] [PubMed]
  76. Jeon, E.S.; Heo, S.C.; Lee, I.H.; Choi, Y.J.; Park, J.H.; Choi, K.U.; Park, D.Y.; Suh, D.-S.; Yoon, M.-S.; Kim, J.H. Ovarian Cancer-Derived Lysophosphatidic Acid Stimulates Secretion of VEGF and Stromal Cell-Derived Factor-1α from Human Mesenchymal Stem Cells. Exp. Mol. Med. 2010, 42, 280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Park, S.Y.; Jeong, K.J.; Panupinthu, N.; Yu, S.; Lee, J.; Han, J.W.; Kim, J.M.; Lee, J.-S.; Kang, J.; Park, C.G.; et al. Lysophosphatidic Acid Augments Human Hepatocellular Carcinoma Cell Invasion through LPA1 Receptor and MMP-9 Expression. Oncogene 2011, 30, 1351–1359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Yu, S.; Sun, L.; Jiao, Y.; Lee, L.T.O. The Role of G Protein-Coupled Receptor Kinases in Cancer. Int. J. Biol. Sci. 2018, 14, 189–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Okabe, K.; Hayashi, M.; Kato, K.; Okumura, M.; Fukui, R.; Honoki, K.; Fukushima, N.; Tsujiuchi, T. Lysophosphatidic Acid Receptor-3 Increases Tumorigenicity and Aggressiveness of Rat Hepatoma RH7777 Cells. Mol. Carcinog. 2013, 52, 247–254. [Google Scholar] [CrossRef]
  80. Zuo, C.; Li, X.; Huang, J.; Chen, D.; Ji, K.; Yang, Y.; Xu, T.; Zhu, D.; Yan, C.; Gao, P. Osteoglycin Attenuates Cardiac Fibrosis by Suppressing Cardiac Myofibroblast Proliferation and Migration through Antagonizing Lysophosphatidic Acid 3/Matrix Metalloproteinase 2/Epidermal Growth Factor Receptor Signalling. Cardiovasc. Res. 2018, 114, 703–712. [Google Scholar] [CrossRef] [Green Version]
  81. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix Metalloproteinases: Regulators of the Tumor Microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Furuta, D.; Yamane, M.; Tsujiuchi, T.; Moriyama, R.; Fukushima, N. Lysophosphatidic Acid Induces Neurite Branch Formation through LPA3. Mol. Cell. Neurosci. 2012, 50, 21–34. [Google Scholar] [CrossRef] [PubMed]
  83. Fukushima, N.; Ishii, S.; Tsujiuchi, T.; Kagawa, N.; Katoh, K. Comparative Analyses of Lysophosphatidic Acid Receptor-Mediated Signaling. Cell. Mol. Life Sci. 2015, 72, 2377–2394. [Google Scholar] [CrossRef]
  84. Chan, L.C.; Peters, W.; Xu, Y.; Chun, J.; Farese, R.V.; Cases, S. LPA3 Receptor Mediates Chemotaxis of Immature Murine Dendritic Cells to Unsaturated Lysophosphatidic Acid (LPA). J. Leukoc. Biol. 2007, 82, 1193–1200. [Google Scholar] [CrossRef] [Green Version]
  85. Zhao, C.; Fernandes, M.J.; Prestwich, G.D.; Turgeon, M.; di Battista, J.; Clair, T.; Poubelle, P.E.; Bourgoin, S.G. Regulation of Lysophosphatidic Acid Receptor Expression and Function in Human Synoviocytes: Implications for Rheumatoid Arthritis? Mol. Pharmacol. 2008, 73, 587–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Nochi, H.; Tomura, H.; Tobo, M.; Tanaka, N.; Sato, K.; Shinozaki, T.; Kobayashi, T.; Takagishi, K.; Ohta, H.; Okajima, F.; et al. Stimulatory Role of Lysophosphatidic Acid in Cyclooxygenase-2 Induction by Synovial Fluid of Patients with Rheumatoid Arthritis in Fibroblast-Like Synovial Cells. J. Immunol. 2008, 181, 5111–5119. [Google Scholar] [CrossRef] [Green Version]
  87. Bandoh, K.; Aoki, J.; Taira, A.; Tsujimoto, M.; Arai, H.; Inoue, K. Lysophosphatidic Acid (LPA) Receptors of the EDG Family Are Differentially Activated by LPA Species. Structure-Activity Relationship of Cloned LPA Receptors. FEBS Lett. 2000, 478, 159–165. [Google Scholar] [CrossRef] [Green Version]
  88. Bandoh, K.; Aoki, J.; Hosono, H.; Kobayashi, S.; Kobayashi, T.; Murakami-Murofushi, K.; Tsujimoto, M.; Arai, H.; Inoue, K. Molecular Cloning and Characterization of a Novel Human G-Protein-Coupled Receptor, EDG7, for Lysophosphatidic Acid. J. Biol. Chem. 1999, 274, 27776–27785. [Google Scholar] [CrossRef] [Green Version]
  89. Alcántara-Hernández, R.; Hernández-Méndez, A.; Campos-Martínez, G.A.; Meizoso-Huesca, A.; García-Sáinz, J.A. Phosphorylation and Internalization of Lysophosphatidic Acid Receptors LPA1, LPA2, and LPA3. PLoS ONE 2015, 10, e0140583. [Google Scholar] [CrossRef] [Green Version]
  90. Fells, J.I.; Tsukahara, R.; Liu, J.; Tigyi, G.; Parrill, A.L. Structure-Based Drug Design Identifies Novel LPA3 Antagonists. Bioorg. Med. Chem. 2009, 17, 7457–7464. [Google Scholar] [CrossRef] [Green Version]
  91. Guo, S.; Zhang, J.; Zhang, S.; Li, J. A Single Amino Acid Mutation (R104P) in the E/DRY Motif of GPR40 Impairs Receptor Function. PLoS ONE 2015, 10, e0141303. [Google Scholar] [CrossRef]
  92. White, K.L.; Eddy, M.T.; Gao, Z.G.; Han, G.W.; Lian, T.; Deary, A.; Patel, N.; Jacobson, K.A.; Katritch, V.; Stevens, R.C. Structural Connection between Activation Microswitch and Allosteric Sodium Site in GPCR Signaling. Structure 2018, 26, 259.e5–269.e5. [Google Scholar] [CrossRef] [Green Version]
  93. Olivella, M.; Caltabiano, G.; Cordomí, A. The Role of Cysteine 6.47 in Class A GPCRs. BMC Struct. Biol. 2013, 13, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kim, Y.-M.; Benovic, J.L. Differential Roles of Arrestin-2 Interaction with Clathrin and Adaptor Protein 2 in G Protein-Coupled Receptor Trafficking. J. Biol. Chem. 2002, 277, 30760–30768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Paing, M.M.; Johnston, C.A.; Siderovski, D.P.; Trejo, J. Clathrin Adaptor AP2 Regulates Thrombin Receptor Constitutive Internalization and Endothelial Cell Resensitization. Mol. Cell. Biol. 2006, 26, 3231–3242. [Google Scholar] [CrossRef] [Green Version]
  96. Wolfe, B.L.; Trejo, J.A. Clathrin-Dependent Mechanisms of G Protein-Coupled Receptor Endocytosis. Traffic 2007, 8, 462–470. [Google Scholar] [CrossRef]
  97. Vickery, O.N.; Carvalheda, C.A.; Zaidi, S.A.; Pisliakov, A.V.; Katritch, V.; Zachariae, U. Intracellular Transfer of Na+ in an Active-State G-Protein-Coupled Receptor. Structure 2018, 26, 171–180. [Google Scholar] [CrossRef] [Green Version]
  98. Rovati, G.E.; Capra, V.; Shaw, V.S.; Malik, R.U.; Sivaramakrishnan, S.; Neubig, R.R. The DRY Motif and the Four Corners of the Cubic Ternary Complex Model. Cell Signal. 2017, 35, 16–23. [Google Scholar] [CrossRef] [PubMed]
  99. Zhou, Q.; Yang, D.; Wu, M.; Guo, Y.; Guo, W.; Zhong, L.; Cai, X.; Dai, A.; Jang, W.; Shakhnovich, E.I.; et al. Common Activation Mechanism of Class A GPCRs. eLife 2019, 8, e50279. [Google Scholar] [CrossRef]
  100. Yuan, S.; Filipek, S.; Palczewski, K.; Vogel, H. Activation of G-Protein-Coupled Receptors Correlates with the Formation of a Continuous Internal Water Pathway. Nat. Commun. 2014, 5, 4733. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, X.C.; Zhou, Y.; Cao, C. Proton Transfer during Class-A GPCR Activation: Do the CWxP Motif and the Membrane Potential Act in Concert? Biophys. Rep. 2018, 4, 115–122. [Google Scholar] [CrossRef] [Green Version]
  102. Moreira, I.S. Structural Features of the G-Protein/GPCR Interactions. Biochim. Biophys. Acta (BBA) 2014, 1840, 16–33. [Google Scholar] [CrossRef] [PubMed]
  103. Wacker, D.; Stevens, R.C.; Roth, B.L. How Ligands Illuminate GPCR Molecular Pharmacology. Cell 2017, 170, 414–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Sutkeviciute, I.; Vilardaga, J.-P. Structural Insights into Emergent Signaling Modes of G Protein–Coupled Receptors. J. Biol. Chem. 2020, 295, 11626–11642. [Google Scholar] [CrossRef] [PubMed]
  105. Ubarretxena-Belandia, I.; Engelman, D.M. Helical Membrane Proteins: Diversity of Functions in the Context of Simple Architecture. Curr. Opin. Struct. Biol. 2001, 11, 370–376. [Google Scholar] [CrossRef]
  106. Alewijnse, A.E.; Timmerman, H.; Jacobs, E.H.; Smit, M.J.; Roovers, E.; Cotecchia, S.; Leurs, R. The Effect of Mutations in the DRY Motif on the Constitutive Activity and Structural Instability of the Histamine H2Receptor. Mol. Pharmacol. 2000, 57, 890–898. [Google Scholar] [PubMed]
  107. He, R.; Browning, D.D.; Ye, R.D. Differential Roles of the NPXXY Motif in Formyl Peptide Receptor Signaling. J. Immunol. 2001, 166, 4099–4105. [Google Scholar] [CrossRef] [Green Version]
  108. Chung, D.A.; Wade, S.M.; Fowler, C.B.; Woods, D.D.; Abada, P.B.; Mosberg, H.I.; Neubig, R.R. Mutagenesis and Peptide Analysis of the DRY Motif in the α2A Adrenergic Receptor: Evidence for Alternate Mechanisms in G Protein-Coupled Receptors. Biochem. Biophys. Res. Commun. 2002, 293, 1233–1241. [Google Scholar] [CrossRef]
  109. Zhou, X.E.; He, Y.; de Waal, P.W.; Gao, X.; Kang, Y.; van Eps, N.; Yin, Y.; Pal, K.; Goswami, D.; White, T.A.; et al. Identification of Phosphorylation Codes for Arrestin Recruitment by G Protein-Coupled Receptors. Cell 2017, 170, 457.e13–469.e13. [Google Scholar] [CrossRef] [Green Version]
  110. Mayer, D.; Damberger, F.F.; Samarasimhareddy, M.; Feldmueller, M.; Vuckovic, Z.; Flock, T.; Bauer, B.; Mutt, E.; Zosel, F.; Allain, F.H.T.; et al. Distinct G Protein-Coupled Receptor Phosphorylation Motifs Modulate Arrestin Affinity and Activation and Global Conformation. Nat. Commun. 2019, 10, 1261. [Google Scholar] [CrossRef]
  111. Fujiwara, Y.; Sardar, V.; Tokumura, A.; Baker, D.; Murakami-Murofushi, K.; Parrill, A.; Tigyi, G. Identification of Residues Responsible for Ligand Recognition and Regioisomeric Selectivity of Lysophosphatidic Acid Receptors Expressed in Mammalian Cells. J. Biol. Chem. 2005, 280, 35038–35050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Valentine, W.J.; Fells, J.I.; Perygin, D.H.; Mujahid, S.; Yokoyama, K.; Fujiwara, Y.; Tsukahara, R.; van Brocklyn, J.R.; Parrill, A.L.; Tigyi, G. Subtype-Specific Residues Involved in Ligand Activation of the Endothelial Differentiation Gene Family Lysophosphatidic Acid Receptors. J. Biol. Chem. 2008, 283, 12175–12187. [Google Scholar] [CrossRef] [Green Version]
  113. Kaye, R.G.; Saldanha, J.W.; Lu, Z.-L.; Hulme, E.C. Helix 8 of the M1 Muscarinic Acetylcholine Receptor: Scanning Mutagenesis Delineates a G Protein Recognition Site. Mol. Pharmacol. 2011, 79, 701–709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Santos, N.M.D.; Gardner, L.A.; White, S.W.; Bahouth, S.W. Characterization of the Residues in Helix 8 of the Human β1-Adrenergic Receptor That Are Involved in Coupling the Receptor to G Proteins. J. Biol. Chem. 2006, 281, 12896–12907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Huynh, J.; Thomas, W.G.; Aguilar, M.I.; Pattenden, L.K. Role of Helix 8 in G Protein-Coupled Receptors Based on Structure-Function Studies on the Type 1 Angiotensin Receptor. Mol. Cell. Endocrinol. 2009, 302, 118–127. [Google Scholar] [CrossRef]
  116. Dijkman, P.M.; Muñoz-García, J.C.; Lavington, S.R.; Kumagai, P.S.; dos Reis, R.I.; Yin, D.; Stansfeld, P.J.; Costa-Filho, A.J.; Watts, A. Conformational Dynamics of a G Protein–Coupled Receptor Helix 8 in Lipid Membranes. Sci. Adv. 2020, 6, eaav8207. [Google Scholar] [CrossRef] [PubMed]
  117. Ribas, C.; Penela, P.; Murga, C.; Salcedo, A.; García-Hoz, C.; Jurado-Pueyo, M.; Aymerich, I.; Mayor, F. The G Protein-Coupled Receptor Kinase (GRK) Interactome: Role of GRKs in GPCR Regulation and Signaling. Biochim. Biophys. Acta BBA Biomembr. 2007, 1768, 913–922. [Google Scholar] [CrossRef] [Green Version]
  118. Gurevich, V.V.; Song, X.; Vishnivetskiy, S.A.; Gurevich, E.V. Enhanced Phosphorylation-Independent Arrestins and Gene Therapy. In Arrestins—Pharmacology and Therapeutic Potential; Gurevich, V.V., Ed.; Springer: Berlin/Heidelberg, Germany, 2014; Volume 219, pp. 133–152. ISBN 978-3-642-41198-4. [Google Scholar]
  119. Pronin, A.N.; Carman, C.V.; Benovic, J.L. Structure-Function Analysis of G Protein-Coupled Receptor Kinase-5. J. Biol. Chem. 1998, 273, 31510–31518. [Google Scholar] [CrossRef] [Green Version]
  120. Watari, K.; Nakaya, M.; Kurose, H. Multiple Functions of G Protein-Coupled Receptor Kinases. J. Mol. Signal. 2014, 9, 1. [Google Scholar] [CrossRef] [Green Version]
  121. Drake, M.T.; Shenoy, S.K.; Lefkowitz, R.J. Trafficking of G Protein-Coupled Receptors. Circ. Res. 2006, 99, 570–582. [Google Scholar] [CrossRef]
  122. Reiter, E.; Lefkowitz, R.J. GRKs and β-Arrestins: Roles in Receptor Silencing, Trafficking and Signaling. Trends Endocrinol. Metab. 2006, 17, 159–165. [Google Scholar] [CrossRef]
  123. Baidya, M.; Kumari, P.; Dwivedi-Agnihotri, H.; Pandey, S.; Chaturvedi, M.; Stepniewski, T.M.; Kawakami, K.; Cao, Y.; Laporte, S.A.; Selent, J.; et al. Key Phosphorylation Sites in GPCR s Orchestrate the Contribution of β-Arrestin 1 in ERK 1/2 Activation. EMBO Rep. 2020, 21, e49886. [Google Scholar] [CrossRef] [PubMed]
  124. Sensoy, O.; Moreira, I.S.; Morra, G. Understanding the Differential Selectivity of Arrestins toward the Phosphorylation State of the Receptor. ACS Chem. Neurosci. 2016, 7, 1212–1224. [Google Scholar] [CrossRef]
  125. Ally, R.A.; Ives, K.L.; Traube, E.; Eltounsi, I.; Chen, P.-W.; Cahill, P.J.; Battey, J.F.; Hellmich, M.R.; Kroog, G.S. Agonist- and Protein Kinase C-Induced Phosphorylation Have Similar Functional Consequences for Gastrin-Releasing Peptide Receptor Signaling via Gq. Mol. Pharmacol. 2003, 64, 890–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Kim, J.; Ahn, S.; Ren, X.-R.; Whalen, E.J.; Reiter, E.; Wei, H.; Lefkowitz, R.J. Functional Antagonism of Different G Protein-Coupled Receptor Kinases for -Arrestin-Mediated Angiotensin II Receptor Signaling. Proc. Natl. Acad. Sci. USA 2005, 102, 1442–1447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Flock, T.; Ravarani, C.N.J.; Sun, D.; Venkatakrishnan, A.J.; Kayikci, M.; Tate, C.G.; Veprintsev, D.B.; Babu, M.M. Universal Allosteric Mechanism for Gα Activation by GPCRs. Nature 2015, 524, 173–179. [Google Scholar] [CrossRef] [Green Version]
  128. Butcher, A.J.; Prihandoko, R.; Kong, K.C.; McWilliams, P.; Edwards, J.M.; Bottrill, A.; Mistry, S.; Tobin, A.B. Differential G-Protein-Coupled Receptor Phosphorylation Provides Evidence for a Signaling Bar Code. J. Biol. Chem. 2011, 286, 11506–11518. [Google Scholar] [CrossRef] [Green Version]
  129. Hernandez-Mendez, A.; Alcantara-Hernandez, R.; Garcia-Sainz, J.A. Lysophosphatidic Acid LPA1-3 Receptors: Signaling, Regulation and in Silico Analysis of Their Putative Phosphorylation Sites. Recept. Clin. Investig. 2014, 1, e193. [Google Scholar] [CrossRef]
  130. Gurevich, V.V.; Gurevich, E.V. GPCR Signaling Regulation: The Role of GRKs and Arrestins. Front. Pharmacol. 2019, 10, 125. [Google Scholar] [CrossRef] [Green Version]
  131. Black, J.B.; Premont, R.T.; Daaka, Y. Feedback Regulation of G Protein-Coupled Receptor Signaling by GRKs and Arrestins. Semin. Cell Dev. Biol. 2016, 50, 95–104. [Google Scholar] [CrossRef] [Green Version]
  132. Lodowski, D.T.; Pitcher, J.A.; Capel, W.D.; Lefkowitz, R.J.; Tesmer, J.J.G. Keeping G Proteins at Bay: A Complex between G Protein-Coupled Receptor Kinase 2 and Gβγ. Science 2003, 300, 1256–1262. [Google Scholar] [CrossRef]
  133. Cassier, E.; Gallay, N.; Bourquard, T.; Claeysen, S.; Bockaert, J.; Crépieux, P.; Poupon, A.; Reiter, E.; Marin, P.; Vandermoere, F. Phosphorylation of β-Arrestin2 at Thr383 by MEK Underlies β-Arrestin-Dependent Activation of Erk1/2 by GPCRs. eLife 2017, 6, e23777. [Google Scholar] [CrossRef]
  134. Wei, H.; Ahn, S.; Shenoy, S.K.; Karnik, S.S.; Hunyady, L.; Luttrell, L.M.; Lefkowitz, R.J. Independent -Arrestin 2 and G Protein-Mediated Pathways for Angiotensin II Activation of Extracellular Signal-Regulated Kinases 1 and 2. Proc. Natl. Acad. Sci. USA 2003, 100, 10782–10787. [Google Scholar] [CrossRef] [Green Version]
  135. Murga, C.; Ruiz-Gómez, A.; García-Higuera, I.; Kim, C.M.; Benovic, J.L.; Mayor, F. High Affinity Binding of β-Adrenergic Receptor Kinase to Microsomal Membranes. J. Biol. Chem. 1996, 271, 985–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Shiina, T.; Arai, K.; Tanabe, S.; Yoshida, N.; Haga, T.; Nagao, T.; Kurose, H. Clathrin Box in G Protein-Coupled Receptor Kinase 2. J. Biol. Chem. 2001, 276, 33019–33026. [Google Scholar] [CrossRef] [Green Version]
  137. Peregrin, S.; Jurado-Pueyo, M.; Campos, P.M.; Sanz-Moreno, V.; Ruiz-Gomez, A.; Crespo, P.; Mayor, F.; Murga, C. Phosphorylation of P38 by GRK2 at the Docking Groove Unveils a Novel Mechanism for Inactivating P38MAPK. Curr. Biol. 2006, 16, 2042–2047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Carman, C.V.; Barak, L.S.; Chen, C.; Liu-Chen, L.Y.; Onorato, J.J.; Kennedy, S.P.; Caron, M.G.; Benovic, J.L. Mutational Analysis of Gβγ and Phospholipid Interaction with Kinase G Protein-Coupled Receptor 2. J. Biol. Chem. 2000, 275, 10443–10452. [Google Scholar] [CrossRef] [Green Version]
  139. Bahouth, S.W.; Nooh, M.M. Barcoding of GPCR Trafficking and Signaling through the Various Trafficking Roadmaps by Compartmentalized Signaling Networks. Cell Signal. 2017, 36, 42–55. [Google Scholar] [CrossRef]
  140. Yang, Z.; Yang, F.; Zhang, D.; Liu, Z.; Lin, A.; Liu, C.; Xiao, P.; Yu, X.; Sun, J.P. Phosphorylation of g Protein-Coupled Receptors: From the Barcode Hypothesis to the Flute Model. Mol. Pharmacol. 2017, 92, 201–210. [Google Scholar] [CrossRef] [PubMed]
  141. Milligan, G. New Aspects of G-Protein-Coupled Receptor Signalling and Regulation. Trends Endocrinol. Metab. 1998, 9, 13–19. [Google Scholar] [CrossRef]
  142. Böttke, T.; Ernicke, S.; Serfling, R.; Ihling, C.; Burda, E.; Gurevich, V.V.; Sinz, A.; Coin, I. Exploring GPCR-arrestin Interfaces with Genetically Encoded Crosslinkers. EMBO Rep. 2020, 21. [Google Scholar] [CrossRef]
  143. Urs, N.M.; Kowalczyk, A.P.; Radhakrishna, H. Different Mechanisms Regulate Lysophosphatidic Acid (LPA)-Dependent versus Phorbol Ester-Dependent Internalization of the LPA1 Receptor. J. Biol. Chem. 2008, 283, 5249–5257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Romero, G.; von Zastrow, M.; Friedman, P.A. Role of PDZ Proteins in Regulating Trafficking, Signaling, and Function of GPCRs: Means, Motif, and Opportunity. In Advances in Pharmacology; Elsevier: Amsterdam, The Netherlands, 2011; Volume 62, pp. 279–314. ISBN 978-0-12-385952-5. [Google Scholar]
  145. Colgan, L.; Liu, H.; Huang, S.Y.; Liu, Y.-J. Dileucine Motif Is Sufficient for Internalization and Synaptic Vesicle Targeting of Vesicular Acetylcholine Transporter. Traffic 2007, 8, 512–522. [Google Scholar] [CrossRef] [PubMed]
  146. Geisler, C.; Dietrich, J.; Nielsen, B.L.; Kastrup, J.; Lauritsen, J.P.H.; Ødum, N.; Christensen, M.D. Leucine-Based Receptor Sorting Motifs Are Dependent on the Spacing Relative to the Plasma Membrane. J. Biol. Chem. 1998, 273, 21316–21323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Moo, E.V.; van Senten, J.R.; Bräuner-Osborne, H.; Møller, T.C. Arrestin-Dependent and -Independent Internalization of G Protein–Coupled Receptors: Methods, Mechanisms, and Implications on Cell Signaling. Mol. Pharmacol. 2021, 99, 242–255. [Google Scholar] [CrossRef] [PubMed]
  148. Novakovic, S.; Sawai, E.T.; Radke, K. Dileucine and YXXL Motifs in the Cytoplasmic Tail of the Bovine Leukemia Virus Transmembrane Envelope Protein Affect Protein Expression on the Cell Surface. J. Virol. 2004, 78, 8301–8311. [Google Scholar] [CrossRef] [Green Version]
  149. Kozik, P.; Francis, R.W.; Seaman, M.N.J.; Robinson, M.S. A Screen for Endocytic Motifs. Traffic 2010, 11, 843–855. [Google Scholar] [CrossRef]
  150. Bonifacino, J.S.; Traub, L.M. Signals for Sorting of Transmembrane Proteins to Endosomes and Lysosomes. Annu. Rev. Biochem. 2003, 72, 395–447. [Google Scholar] [CrossRef] [Green Version]
  151. Pandey, K.N. Small Peptide Recognition Sequence for Intracellular Sorting. Curr. Opin. Biotechnol. 2010, 21, 611–620. [Google Scholar] [CrossRef] [Green Version]
  152. Christensen, N.R.; Čalyševa, J.; Fernandes, E.F.A.; Lüchow, S.; Clemmensen, L.S.; Haugaard-Kedström, L.M.; Strømgaard, K. PDZ Domains as Drug Targets. Adv. Ther. 2019, 2, 1800143. [Google Scholar] [CrossRef] [Green Version]
  153. Varsano, T.; Taupin, V.; Guo, L.; Baterina, O.Y.; Farquhar, M.G. The PDZ Protein GIPC Regulates Trafficking of the LPA1 Receptor from APPL Signaling Endosomes and Attenuates the Cell’s Response to LPA. PLoS ONE 2012, 7, e49227. [Google Scholar] [CrossRef] [Green Version]
  154. Holcomb, J.; Jiang, Y.; Lu, G.; Trescott, L.; Brunzelle, J.; Sirinupong, N.; Li, C.; Naren, A.P.; Yang, Z. Structural Insights into PDZ-Mediated Interaction of NHERF2 and LPA2, a Cellular Event Implicated in CFTR Channel Regulation. Biochem. Biophys. Res. Commun. 2014, 446, 399–403. [Google Scholar] [CrossRef] [Green Version]
  155. Zhang, H.; Wang, D.; Sun, H.; Hall, R.A.; Yun, C.C. MAGI-3 Regulates LPA-Induced Activation of Erk and RhoA. Cell Signal. 2007, 19, 261–268. [Google Scholar] [CrossRef] [Green Version]
Figure 1. LPA structure. Chemical structure of 1-oleoyl-2-hydroxy-sn-glycerol-3-phosphate (LPA 18:1). Atoms in the chemical structure: Carbon (grey), Hydrogen (white), Oxygen (red), and Phosphorus (orange) (https://pubchem.ncbi.nlm.nih.gov/compound/Lysophosphatidic-acid) (https://molview.org). Accessed on 4 June 2021.
Figure 1. LPA structure. Chemical structure of 1-oleoyl-2-hydroxy-sn-glycerol-3-phosphate (LPA 18:1). Atoms in the chemical structure: Carbon (grey), Hydrogen (white), Oxygen (red), and Phosphorus (orange) (https://pubchem.ncbi.nlm.nih.gov/compound/Lysophosphatidic-acid) (https://molview.org). Accessed on 4 June 2021.
Ijms 22 06704 g001
Figure 2. LPA receptors and G proteins. LPA receptors couple with different G proteins that activate distinct signaling pathways. PLC, phospholipase C; PI3K. phosphoinositide 3-kinase; AC, adenylyl cyclase. Created with BioRender.com.
Figure 2. LPA receptors and G proteins. LPA receptors couple with different G proteins that activate distinct signaling pathways. PLC, phospholipase C; PI3K. phosphoinositide 3-kinase; AC, adenylyl cyclase. Created with BioRender.com.
Ijms 22 06704 g002
Figure 3. Internalization of agonist-activated LPA3 receptors. (1) Activation of LPA3 with LPA and recruitment of a G protein. (2) Exposure of GPCR phosphorylation sites. (3) Recruitment of β-arrestin through interaction with phosphorylated sites. (4) Recruitment of the endocytic machinery that initiates receptor endocytosis. (5) Endocytosis of LPA3 via endosomes. (6) Receptor-endosomal traffic to (7) lysosomal receptor degradation or (8) receptor recycling to the plasma membrane. Question marks indicate that there is little information on these processes, which are postulated in similarity to what has been defined for other receptors. Created with BioRender.
Figure 3. Internalization of agonist-activated LPA3 receptors. (1) Activation of LPA3 with LPA and recruitment of a G protein. (2) Exposure of GPCR phosphorylation sites. (3) Recruitment of β-arrestin through interaction with phosphorylated sites. (4) Recruitment of the endocytic machinery that initiates receptor endocytosis. (5) Endocytosis of LPA3 via endosomes. (6) Receptor-endosomal traffic to (7) lysosomal receptor degradation or (8) receptor recycling to the plasma membrane. Question marks indicate that there is little information on these processes, which are postulated in similarity to what has been defined for other receptors. Created with BioRender.
Ijms 22 06704 g003
Figure 4. Signaling pathway of LPA3 receptors. Activation of this receptor subtype with LPA promotes conformational changes favoring intense interaction with Gαq/11 and Gαi/o, which lead to activation of downstream signaling molecular entities. Abbreviations as in Figure 2. Created with BioRender.
Figure 4. Signaling pathway of LPA3 receptors. Activation of this receptor subtype with LPA promotes conformational changes favoring intense interaction with Gαq/11 and Gαi/o, which lead to activation of downstream signaling molecular entities. Abbreviations as in Figure 2. Created with BioRender.
Ijms 22 06704 g004
Figure 5. LPA3 receptor structure, domains and sites that regulate this receptor. Image shows the amino acid sequence and the organization of the LPA3 receptor with three extracellular loops, three intracellular loops, the seven transmembrane domains, the extracellular amino terminus (-NH2), and the intracellular carboxyl terminus (-COOH). Colored boxes indicate conserved motifs putatively relevant for activation and regulation of the LPA3 receptor. Putative sites where LPA interacts with LPA3 are shown in green, while proposed places where GPCRs could be recruiting G proteins are marked in blue and purple (R, arginine that is also part of the ERH motif). “Y” indicates a potential glycosylation site, and the line joining one of the cysteines to the membrane is a putative palmitoylation site.
Figure 5. LPA3 receptor structure, domains and sites that regulate this receptor. Image shows the amino acid sequence and the organization of the LPA3 receptor with three extracellular loops, three intracellular loops, the seven transmembrane domains, the extracellular amino terminus (-NH2), and the intracellular carboxyl terminus (-COOH). Colored boxes indicate conserved motifs putatively relevant for activation and regulation of the LPA3 receptor. Putative sites where LPA interacts with LPA3 are shown in green, while proposed places where GPCRs could be recruiting G proteins are marked in blue and purple (R, arginine that is also part of the ERH motif). “Y” indicates a potential glycosylation site, and the line joining one of the cysteines to the membrane is a putative palmitoylation site.
Ijms 22 06704 g005
Figure 6. In silico prediction of serine and threonine sites phosphorylated by GRK, PKA and PKC. LPA3 structure is represented, showing (in red) the putative sites targeted by GRK and (in cerulean) putative sites phosphorylated by PKA or PKC.
Figure 6. In silico prediction of serine and threonine sites phosphorylated by GRK, PKA and PKC. LPA3 structure is represented, showing (in red) the putative sites targeted by GRK and (in cerulean) putative sites phosphorylated by PKA or PKC.
Ijms 22 06704 g006
Table 1. GRKs that putatively phosphorylate different sites in GPCRs.
Table 1. GRKs that putatively phosphorylate different sites in GPCRs.
SubfamiliesGRKsDomains of Interest
Visual GRKsGRK1 and GRK7Prenylation
GRK2 or βARK
GRK4
GRK2 and GRK3
GRK4, GRK5 and GRK6
Pleckstrin homology
Palmitoylation, polybasic hydrophobic domains
Table 2. In silico prediction of PKC and PKA phosphorylated LPA3 residues.
Table 2. In silico prediction of PKC and PKA phosphorylated LPA3 residues.
PositionAmino AcidPKC/PKA
130SPKA
217TPKCα/PKCδ/PKCγ
233TPKA/PKCδ/PKCι/PKCζ
243TPKCi/PKCζ
321SPKA/PKCδ/
325SPKA/PKC/PKCε
341SPKCε
351SPKCε
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Solís, K.H.; Romero-Ávila, M.T.; Guzmán-Silva, A.; García-Sáinz, J.A. The LPA3 Receptor: Regulation and Activation of Signaling Pathways. Int. J. Mol. Sci. 2021, 22, 6704. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136704

AMA Style

Solís KH, Romero-Ávila MT, Guzmán-Silva A, García-Sáinz JA. The LPA3 Receptor: Regulation and Activation of Signaling Pathways. International Journal of Molecular Sciences. 2021; 22(13):6704. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136704

Chicago/Turabian Style

Solís, Karina Helivier, M. Teresa Romero-Ávila, Alejandro Guzmán-Silva, and J. Adolfo García-Sáinz. 2021. "The LPA3 Receptor: Regulation and Activation of Signaling Pathways" International Journal of Molecular Sciences 22, no. 13: 6704. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22136704

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop