• Open Access
    Review

    Lysophospholipid receptors in neurodegeneration and neuroprotection

    Eric Birgbauer *

    Explor Neuroprot Ther. 2024;4:349–365 DOI: https://doi.org/10.37349/ent.2024.00088

    Received: April 16, 2024 Accepted: August 11, 2024 Published: August 22, 2024

    Academic Editor: Dasiel O. Borroto Escuela, Karolinska Institutet, Sweden; Rafael Franco, Universidad de Barcelona, Spain

    This article belongs to the special issue GPCR Heteroreceptor Complexes as Key Players in Neuroprotection

    Abstract

    The central nervous system (CNS) is one of the most complex physiological systems, and treatment of CNS disorders represents an area of major medical need. One critical aspect of the CNS is its lack of regeneration, such that damage is often permanent. The damage often leads to neurodegeneration, and so strategies for neuroprotection could lead to major medical advances. The G protein-coupled receptor (GPCR) family is one of the major receptor classes, and they have been successfully targeted clinically. One class of GPCRs is those activated by bioactive lysophospholipids as ligands, especially sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA). Research has been increasingly demonstrating the important roles that S1P and LPA, and their receptors, play in physiology and disease. In this review, I describe the role of S1P and LPA receptors in neurodegeneration and potential roles in neuroprotection. Much of our understanding of the role of S1P receptors has been through pharmacological tools. One such tool, fingolimod (also known as FTY720), which is a S1P receptor agonist but a functional antagonist in the immune system, is clinically efficacious in multiple sclerosis by producing a lymphopenia to reduce autoimmune attacks; however, there is evidence that fingolimod is also neuroprotective. Furthermore, fingolimod is neuroprotective in many other neuropathologies, including stroke, Parkinson’s disease, Huntington’s disease, Rett syndrome, Alzheimer’s disease, and others that are discussed here. LPA receptors also appear to be involved, being upregulated in a variety of neuropathologies. Antagonists or mutations of LPA receptors, especially LPA1, are neuroprotective in a variety of conditions, including cortical development, traumatic brain injury, spinal cord injury, stroke and others discussed here. Finally, LPA receptors may interact with other receptors, including a functional interaction with plasticity related genes.

    Keywords

    Sphingosine-1-phosphate, S1P receptors, lysophosphatidic acid, LPA receptors, Lpar, S1pr, fingolimod, FTY720

    Introduction

    G protein-coupled receptors (GPCRs) are one of the largest classes of receptors, with an estimated 800 in the human genome (excluding olfactory receptors). They have a large and diverse set of ligands. Furthermore, they have been the target of many pharmacological compounds in use today [1].

    One class of GPCR ligands is the bioactive lysophospholipids, specifically lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). These lysophospholipid molecules are released from cells and bind to specific receptors, whereby they mediate various cellular responses. These receptors are classic GPCRs that signal through canonical G protein signal transduction pathways. There are six known LPA receptors (LPA1 through LPA6) and five known receptors for S1P (S1P1 through S1P5), each signaling through one or more G proteins (Table 1) [2, 3]. The S1P1 receptor is unique in that all evidence to date suggests that it only signals through Gi/o, where it can decrease adenylate cyclase activity, as well as stimulate the rat sarcoma (Ras) and extracellular signal-regulated kinase (ERK) pathway to enhance cellular proliferation and activate the phosphatidylinositol-3 kinase (PI3K) pathway to inhibit apoptosis. The Gi/o pathway can also activate phospholipase C (PLC) and protein kinase C to increase intracellular calcium. The receptors S1P4 and S1P5 signal through G12/13 to activate Rho in addition to signaling through Gi/o, with some evidence that they may signal through Gs under certain circumstances. The receptors S1P2 and S1P3 signal not only through Gi/o and G12/13, but also through Gq, where they can activate PLC to increase intracellular calcium.

    Lysophospholipid G protein-coupled receptors (GPCR). Nomenclature of the GPCRs and genes of S1P and LPA receptors as well as the G protein cell signaling pathways activated [2, 3]

    LigandGPCR (protein)Gene name
    Mouse/human
    Other namesSignaling pathways
    S1PS1P1S1pr1/S1PR1edg1, lpB1Gi/o
    S1P2S1pr2/S1PR2edg5, lpB2, AGR16, H218Gi/o, Gq, G12/13
    S1P3S1pr3/S1PR3edg3, lpB3Gi/o, Gq, G12/13
    S1P4S1pr4/S1PR4edg6, lpC1Gi/o, G12/13, (Gs)
    S1P5S1pr5/S1PR5edg8, lpB4, Nrg-1Gi/o, G12/13, (Gs)
    LPALPA1Lpar1/LPAR1vzg1, edg2, lpA1Gi/o, Gq, G12/13
    LPA2Lpar2/LPAR2edg4, lpA2Gi/o, Gq, G12/13
    LPA3Lpar3/LPAR3edg7, lpA3Gi/o, Gq
    LPA4Lpar4/LPAR4GPR23, p2y9Gs, Gq, G12/13
    LPA5Lpar5/LPAR5GPR92Gq, G12/13
    LPA6Lpar6/LPAR6p2y5Gs, G12/13
    Display full size

    S1P4 and S1P5 primarily signal through Gi/o and G12/13, although there is some suggestion that they can signal through Gs (parentheses) under certain circumstances

    LPA receptors, likewise, signal through various G protein pathways. The first two receptors identified, LPA1 and LPA2, signal through Gi/o, Gq, and G12/13, with the G12/13 pathway leading to activation of Rho and cytoskeletal rearrangements. The receptor LPA3 signals through Gi/o and Gq, but not G12/13. LPA4 signals through Gq and G12/13, as well as Gs (but not Gi/o). LPA5, on the other hand, only signals through Gq, and G12/13. The final validated LPA receptor, LPA6, which appears to be a lower affinity receptor [4], has been shown to signal through Gs and G12/13.

    LPA and S1P receptors and their ligands have been found to be involved in a variety of physiological and pathological responses (for review, see [5]). This includes extensive roles in the nervous system and neural development (the reader is referred to other reviews [3, 6]). Activation of S1P or LPA receptors is generally considered pro-proliferative and anti-apoptotic, and thus there is considerable research on their role in the progression of cancer [7]. Furthermore, both S1P and LPA receptors are involved in inflammation, including neuroinflammation, and this is another area of active research. There is also evidence of a role of S1P receptors in the cardiovascular system and angiogenesis. In this review, I focus on summarizing evidence that LPA and S1P receptors are involved in neurodegeneration and neuroprotection.

    S1P receptors

    S1P receptors are expressed throughout development and in the adult in many tissues, including the brain and spinal cord. The receptors S1P1 and S1P2 are widely expressed throughout the body [8, 9]. There is major expression in the heart for receptors S1P1, S1P3, and S1P5 [8]. The receptors S1P2 and S1P3 are expressed during development in mesenchymal tissue and somites [8]. S1P4 is highly expressed in the immune system. In the CNS, the receptors S1P1, S1P2, S1P3, and S1P5 are highly expressed [8, 9]. In the brain, expression of S1P1 and S1P4 is found in forebrain. The receptor S1P5, however, is primarily restricted to the brain and spinal cord, where it is highly expressed in oligodendrocytes [8, 1012]. The receptors S1P1 and S1P2 are also expressed during development in neural progenitors in the ventricular zone as well as the neural tube.

    Much of our understanding of the role of S1P receptors in neuropathological conditions and neuroprotection has been elucidated with various pharmacological agents (Table 2). The most widely used pharmacological tool has been the compound fingolimod (also known as FTY720, Gilenya®), which is currently in clinical use for multiple sclerosis (MS). It is an agonist for all the S1P receptors except S1P2, although it often functions as a functional antagonist (see below). However, more specific S1P receptor agonists have been developed, with the S1P1-specific agonist SEW2871 being used more recently.

    Pharmacological compounds. Major pharmacological compounds used in the literature for investigation of the roles of S1P and LPA receptors

    CompoundReceptor targetsRoleReference
    S1P receptor targeting agents
    Fingolimod*S1P1, S1P3, S1P4, S1P5Agonist (functional antagonist)[13, 14]
    SEW2871S1P1Agonist[15, 16]
    W146S1P1Antagonist[17]
    LPA receptor targeting agents
    Ki16425LPA1, LPA3Antagonist[18]
    AM095LPA1Antagonist[19]
    TCLPA5LPA5Antagonist[20]
    Display full size

    * Fingolimod is phosphorylated in vivo, and fingolimod-phosphate is the active agonist. Furthermore, although fingolimod is a S1P receptor agonist, it often acts as a functional antagonist by causing receptor internalization and degradation [21, 22]

    With the approval of fingolimod for the treatment of MS, MS is one of the best examples of a role for S1P receptors in neurological disease. MS is considered an autoimmune disease in which T-lymphocytes cross the blood-brain barrier and attack myelin, leading to demyelinated lesions. In the most common form, relapsing-remitting MS (RR-MS), this demyelination is repaired by oligodendrocytes, most likely newly differentiated from resident oligodendrocyte precursor cells. However, the cycle continues and leads to progressive neuronal damage, and disability, as the disease progresses.

    S1P receptors, especially S1P1, are important clinical targets for MS. As mentioned previously, the pharmacological compound fingolimod has been approved by the FDA for the treatment of RR-MS after clinical trials [23], with many other compounds in clinical trials [3, 21, 24]. Fingolimod (originally called FTY720) was synthesized as a derivative of the fungal metabolite myriocin (also known as ISP-1) as an immunosuppressant for organ graft survival [2527]. Interestingly, it led to lymphopenia with reduced circulating lymphocytes [27]. Later it was discovered that fingolimod is phosphorylated in vivo, and the phosphorylated form is the active compound [13, 14]. Phosphorylated fingolimod has structural similarities to S1P, and it was subsequently shown that fingolimod-phosphate binds to the S1P receptors S1P1, S1P4, and S1P5 with high affinity (EC50 ~0.3–0.6 nM) and to S1P3 with slightly lower affinity (EC50 ~3 nM), but not to S1P2 [13, 14]. Binding of fingolimod to the S1P receptor activates it, and fingolimod was initially characterized as an agonist [13, 14]. However, in the immune system, upon binding fingolimod, S1P receptors are internalized and degraded so that they are unresponsive to S1P, and fingolimod is now often considered a functional antagonist (see [13, 21, 22]). Recently, more specific S1P receptor agonists and antagonists have been developed, especially for S1P1, and many are in various stages of clinical trials [3]. One of the most important ones for experimental investigation has been SEW2871, which is specific for S1P1 (see Table 2).

    Fingolimod’s approval for RR-MS was after extensive preclinical work where it was efficacious in the rodent model experimental autoimmune encephalomyelitis (EAE) [13, 21, 22, 28]. In MS and EAE, fingolimod works by inducing a lymphopenia of peripheral lymphocytes, including CD4+ T cells, CD8+ T cells and B cells. This lymphopenia results in sequestration of central memory T lymphocytes in lymph nodes, reducing the autoimmune activity in MS and EAE.

    However, in addition to immune modulation, fingolimod has been suggested to have a neuroprotective role in EAE and potentially MS [22]. One piece of evidence for this neuroprotective role is that a higher dose of fingolimod is required for efficacy to reduce symptoms in EAE than is needed for lymphocyte sequestration [29]. Furthermore, fingolimod has been shown to restore electrophysiological function after EAE [30]. In addition, fingolimod may also work by reducing neuroinflammation through astrocytes and/or microglia [22, 31].

    In addition to MS, a neuroprotective role for fingolimod has been suggested for many other neurological diseases [3, 32]. For instance, fingolimod is neuroprotective in a cerebral ischemia (stroke) model. During a stroke, a blood clot forms in the brain leading to local ischemia, and the resulting loss of oxygen results in neuronal death; it also produces a neuroinflammatory state, which, especially after reperfusion, can induce further apoptosis. S1P is released after cerebral ischemia in a rodent model, which leads to increased damage. The released S1P was shown to activate S1P receptors on microglia leading to neuroinflammation; there was also some evidence of activation of astrocytes leading to astrogliosis. This neuroinflammation led to neurodegeneration, and fingolimod was neuroprotective but not directly on neurons. Various studies suggest that the receptors S1P1, S1P2, and S1P3 are involved in mediating this response [33], although fingolimod likely acts primarily through S1P1 as its action is mimicked by SEW2871. On the other hand, in a different stroke model, it was found that neural progenitor cells migrated toward a brain infarction, and this was dependent on the receptors S1P1 and S1P2 [34], suggesting a role in neural repair processes.

    There is also evidence that S1P receptor activation is neuroprotective in a Parkinson’s disease model. Parkinson’s disease is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. Furthermore, in two different mouse models of Parkinson’s disease (6-OHDA and rotenone), fingolimod was neuroprotective for dopaminergic neuron loss and apoptosis, as well as reducing dopamine loss and improving motor deficits [35, 36]. This effect appeared direct, as fingolimod treatment also reduced apoptosis by 6-OHDA or rotenone in vitro in the dopaminergic SH-SY5Y cell line. In this model, fingolimod appears to act as a S1P receptor agonist, as its neuroprotective effect is blocked by a S1P1 receptor antagonist (W146). Fingolimod, as well as the S1P1 receptor agonist SEW2871, is also neuroprotective against an in vitro oxidative stress model in these SH-SY5Y cells [37], again showing a role in protecting dopaminergic neurons.

    In yet another neurodegenerative disease, Huntington’s disease, fingolimod was neuroprotective, leading to reduced apoptosis as well as improvements in motor skills, electrophysiology, and survival [38]. This was also recapitulated in a striatal-derived cell line, suggesting a direct neuroprotective effect on neurons.

    In amyotrophic lateral sclerosis (ALS), fingolimod slowed disease progression in a mouse model, likely moderating neuroinflammation, although there may be a direct neuroprotective effect [39]. Furthermore, in vitro, fingolimod is neuroprotective for neurons in culture as well as possibly astrocytes and oligodendrocytes [22].

    A potential mechanism of neuroprotection by S1P receptors is through increasing brain derived neurotrophic factor (BDNF) expression (Figure 1). BDNF is a neurotrophin that is a survival factor for many neurons as well as their axons and synaptic connections. In a mouse model for Rett syndrome, which has low levels of BDNF, fingolimod treatment raised brain BDNF levels as well as promoted improved motor function and increased survival [40]. The authors then went on to show that in an in vitro cortical neuron culture model, either fingolimod, S1P, or the S1P1 receptor agonist SEW2871 increased excitatory neuronal activity. This was accompanied by increased phospho-ERK and phospho-CREB (cAMP-response element binding protein), resulting in increased BDNF production. This increased BDNF production was neuroprotective against excitotoxicity induced by the glutamate agonist N-methyl-D-aspartate (NMDA). This proposed neuroprotective mechanism of increasing BDNF through S1P receptor signaling is also supported in the Huntington disease model, where fingolimod treatment leads to increased BDNF levels as well as being efficacious in improving disease symptoms [38].

    Model for neuroprotection by S1P1 receptors through BDNF. It has been found that activation of S1P1 by S1P leads to increased excitatory neural activity in cortical neurons. Activation of S1P1 as well as neural activity leads to phosphorylation of ERK (pERK). This in turn leads to phosphorylation of cAMP-response element binding protein (pCREB) that results in increased production of brain derived neurotrophic factor (BDNF). BDNF is released and binds to tyrosine receptor kinase B (TrkB) to mediate neuroprotection. BDNF could activate TrkB in the same cell (left in figure) or another cell (right in figure) (Model based upon data from [40])

    Another area where S1P receptors are involved in neuroprotection is in spinal cord injury, which is a complex injury resulting in neuroinflammation, scar formation, axonal loss, and neuronal death. S1P is produced by microglia and astrocytes at the site of injury after a spinal cord lesion [41]. After a contusion spinal lesion, neural stem cells migrate to the site of injury, and this migration is blocked by a short hairpin RNA (shRNA) or inhibitor of S1P1 [41]. Furthermore, fingolimod is neuroprotective in a contusion spinal cord injury model, leading to improvements in clinical score and motor coordination [42]. Interestingly, fingolimod did not reduce inflammation, suggesting a more direct neuroprotective role, although it did reduce astrocyte accumulation. This improvement was recapitulated by the S1P1 receptor specific compound SEW2871, implicating S1P1 as the important receptor.

    S1P receptors also appear to be neuroprotective in Alzheimer’s disease. S1P was found to be increased in the plasma of Alzheimer’s disease patients [43], although this is a correlation and thus may be a result, not a cause, of neuroinflammation. In rat models of Alzheimer’s disease, intracerebroventricular injection of Aβ1-42 leads to hippocampal cell apoptosis and impairments in learning and memory. In this model, treatment with fingolimod was neuroprotective for hippocampal cell loss [44, 45] as well as improving learning and memory in various paradigms [4446]. Intriguingly, in one study, treatment with fingolimod itself impaired spatial learning and memory in control rats [44], suggesting more complex regulation, which may involve multiple S1P receptors. Furthermore, as before, BDNF may be involved. In the model of intracerebroventricular injection of oligomeric Aβ1-42, treatment with fingolimod was neuroprotective and also correlated with increased levels of BDNF [46]. The role of BDNF was corroborated in an in vitro system of oligomeric Aβ1-42 toxicity of cortical neurons, where fingolimod was shown to be neuroprotective, and this neuroprotection was shown to depend on increased BDNF production and signaling through TrkB receptors and ERK1/2 activation, as determined through the use of inhibitors [47]. A more specific role for the receptor S1P1 was seen in a hippocampal slice culture system where the specific agonist SEW2871 significantly reduced tau phosphorylation [48], another hallmark of Alzheimer’s disease.

    The role of S1P receptors has been investigated in other neurological diseases as well. In a mouse model of the neurodegenerative disorder Sandhoff disease, a neuropathic lysosomal storage disorder, progression of the disease involved the receptor S1P3, as mice with null mutations in S1P3 had reduced disease progression and severity, although they were not cured [49]. This progression also required sphingosine kinase 1 (SphK1), the enzyme that catalyzes production of S1P, as a null mutation in SphK1 also reduced disease progression. In this case, the mutations were neuroprotective by reducing astrocyte proliferation and astrogliosis.

    In the eye, in a rat model of retinal disease, S1P appears to be involved in retinal ganglion cell (RGC) death by glutamate excitotoxicity [50]. A sphingosine kinase inhibitor was neuroprotective and reduced the cell death, although the specific S1P receptors were not determined.

    Additional human studies suggest a role for S1P receptors in a variety of neurodegenerative diseases, although this is correlative and specific receptors have not been identified. In human patients, plasma S1P levels were decreased in a variety of neurodegenerative diseases, including idiopathic Parkinson’s disease, Alzheimer’s disease (mentioned above), dementia with Lewy bodies, multiple system atrophy, and progressive supranuclear palsy [43].

    Other studies have investigated the role of S1P receptors in neuroprotection in vitro. In one of the earlier studies, S1P reduced apoptosis caused by serum withdrawal in PC12 cells, a peripheral neural cell line [51], although it was not clear if this was receptor mediated. In an in vitro model of glutamate excitotoxicity in mesencephalic neurons, S1P was neuroprotective [52], but again the receptors involved were not analyzed. In astrocytes, the receptors S1P2 and S1P3 activate neuroinflammation [53].

    Thus, in a variety of model systems, S1P and S1P receptors have neuroprotective roles, although whether their activation or inhibition is neuroprotective varies. Much of this hinges on whether the pharmacological compound fingolimod, as well as SEW2871, acts as an agonist or a functional antagonist, with some evidence for each role.

    LPA receptors

    The roles of LPA receptors in neural development, physiology, and pathogenesis are numerous (for reviews, see [5, 6, 9, 54]). For instance, LPA receptors, especially LPA1 and LPA2, are involved in the early development of the cerebral cortex, regulating neural progenitor expansion and differentiation (see below). A significant role for LPA receptors, in this case, LPA1, LPA3, and LPA5, has been described in the initiation and maintenance of neuropathic pain (see [6, 55, 56]), whereby initiation of neuropathic pain involves a feed-forward mechanism by LPA through LPA1 and LPA3 on microglia. Subsequently, LPA5 is required for maintenance of the neuropathic pain state [57]. Additional roles for LPA receptors, especially LPA1, have been demonstrated in maturation of glutamatergic synapses in the hippocampus, with learning and memory deficits seen in Lpar1 null mice [6, 58].

    LPA receptors are expressed throughout the nervous system, especially during development, with different expression patterns for the six LPA receptors. The receptor genes Lpar1, Lpar2, Lpar4, and Lpar6 are expressed during embryonic brain development in the neocortex, hippocampus, cerebellum, and olfactory bulb, as well as in adult brain in these regions. The receptor Lpar3, however, is only expressed postnatally, and Lpar5 expression may be in early brain development, although the data is less clear [59, 60].

    Outside of the nervous system, LPA receptors have significant roles in cancer, the immune system, as well as other physiological processes (for reviews, see [7, 9, 54, 6166]). Due to the importance of LPA receptors in physiology and disease, various pharmacological compounds are being developed both as experimental tools and for clinical therapeutic purposes (see [63, 67]). However, the LPA receptor pharmacological compounds have not advanced as much as for S1P receptors, and none are yet approved for clinical treatment. Nevertheless, there are a few pharmacological tools that have been used in various studies, especially that inhibit LPA1 (Table 2). The compound Ki16425 has been shown to inhibit LPA1, but also LPA3, and so is not completely specific [18]. A newer, more specific inhibitor of LPA1, AM095, is now being used [19]. In addition, a partially characterized inhibitor of LPA5, TCLPA5, has been used in some studies [20]. However, without good pharmacological agents, much of our understanding of LPA receptor function has been from mice with LPA receptor mutations.

    Focusing on neuroprotection, one of the early roles of LPA receptors is in the development of the cortex. In an in vitro embryonic cortical culture system, LPA addition increases survival of neural progenitors, which is blocked by double null mutations in the receptors LPA1 and LPA2 [68]. Furthermore, in a LPA1 (receptor) null mouse (maLPA1), there was reduced neural progenitor proliferation and increased apoptosis, leading to reduced cortical layers [69]. This suggests a neuroproliferative and/or neuroprotective role of the receptor LPA1. Furthermore, in the adult hippocampus, the maLPA1 null mutant mouse had reduced hippocampal neurogenesis compared to control when exposed to an enriched environment [70], documenting a role for the receptor LPA1 in adult neurogenesis. Indeed, Lpar1 null mice have deficits in learning and memory [71, 72]; whether this was due to a neuroprotective role for LPA1 or a role in synaptic remodeling or neurophysiology is not clear.

    LPA and LPA receptors have been implicated in embryonic axonal growth and, potentially, in axon guidance. LPA has been demonstrated in vitro to cause the collapse of axonal growth cones and neurite retraction in neuroblastoma and PC12 peripheral nerve cell lines [73, 74] as well as primary neurons in culture [7579]. Although the GPCRs mediating these responses have not been identified yet (but see [78]), the response appears receptor mediated due to specificity and potency [78, 79]. This growth cone collapse and neurite retraction proceeds via the Gα12/13 signal transduction pathway, as it is blocked by inhibition of Rho and ROCK [77, 7986]. There also appears to be a role for the receptor LPA3 in neurite branching through the novel GTPase Rnd2 [87].

    In addition to roles in neural development, LPA receptors have been implicated in a variety of neurodegenerative conditions. In traumatic brain injury (TBI), LPAR2 gene expression was increased in human patients ~43 hours (range: 6 hours to 122 hours) after injury [88]. Furthermore, in human patients, levels of the ligand LPA increased significantly 24 hours after injury [89]. Using a mouse model of TBI, treatment with a monoclonal antibody against LPA (Lpathomab) was neuroprotective, leading to reduced lesion volume and decreased cytokine levels, as well as improved behavioral outcomes [89].

    There has also been found a neuroprotective role of LPA receptor antagonism in spinal cord injury. In a mouse model of spinal cord injury, the receptor genes Lpar2 and Lpar3 were upregulated after injury [90]. Spinal cord injury leads to demyelination, but this demyelination was partially blocked in an Lpar1 null mutation mouse or upon treatment with the LPA1 antagonist AM095 [91]. The antagonist also led to a small, but significant, functional improvement. The receptor LPA2 also appears to be involved, as the Lpar2 null mutant mouse also showed partially reduced demyelination and some functional recovery [92]. In another study with a different model of spinal cord injury, an LPA1 antagonist led to increased corticospinal tract sprouting after injury [93], again demonstrating a neuroprotective role by blocking LPA1. Thus, in spinal cord injury, it appears that antagonism of LPA receptors is neuroprotective.

    In another neurodegenerative condition, stroke and cerebral ischemia, LPA acting through LPA receptors appears to mediate neurological damage, and again antagonism is neuroprotective. In human stroke patients, plasma LPA levels increase [94, 95]; LPA levels also increase in the brain in a rodent model of stroke, a transient middle cerebral artery occlusion (tMCAO), which produces a transient focal cerebral ischemia [96, 97]. In this tMCAO model, increased LPA appears to be pathological, as addition of exogenous LPA leads to increased lesion volume [98, 99]. The receptor LPA1 appears to be involved in mediating this pathway, as the antagonist AM095 or an shRNA was neuroprotective and reduced lesion volume, neuronal apoptosis, and neurological deficits after tMCAO and reperfusion [100]. Furthermore, a role for LPA5 has also been implicated by treatment with an antagonist, TCLPA5, after tMCAO and reperfusion leading to functional recovery and reduced lesion volume [101]. Although the specificity of TCLPA5 has not been extensively tested, it inhibits LPA5 [20]. Importantly, treatment with TCLPA5 three hours after cerebral ischemia and reperfusion was neuroprotective, suggesting potential clinical benefit [101]. However, whether the neuroprotective effect is direct on neurons is not clear, as cerebral ischemia also induces neuroinflammation, and an antagonist of LPA1 or LPA5 reduces microglial activation and proinflammatory cytokines [100103]. Interestingly, it was found that when rats are treated with a repeated hyperbaric oxygen exposure prior to ischemia, which is neuroprotective, Lpar1 gene expression was increased [104].

    There is a suggestion for a role of LPA receptors in Alzheimer’s disease pathology (see [105, 106]). However, much of it is indirect, whereby characteristics of Alzheimer’s disease are mimicked by LPA or LPA receptors in other systems, sometimes with extremely high levels of LPA. For instance, LPA treatment of neuroblastoma cells leads to tau phosphorylation, which is a hallmark of Alzheimer’s disease [107]. More recently, there have been found differences in LPA receptor expression in the brain of a transgenic Alzheimer’s disease mouse model [108], but the role of these differences is not clear.

    There is a potential role for receptor LPA2 in ALS. In human ALS patients, there are increased levels of Lpar2 mRNA in the spinal cord, which is also seen in the SOD1G93A mouse model of ALS [109]. Furthermore, in this mouse model, a null mutation in Lpar2 leads to reduced disease progression, suggesting LPA2 is involved in disease mediation. Interestingly, though, the Lpar2 null mutation decreases survival in this mouse model. Thus, in this ALS model, although signaling through LPA2 increases disease symptoms, it extends lifespan. Furthermore, the role of LPA2 appears to be related to inflammation and not directly on motor neurons.

    There is also evidence of the role of LPA receptors in other neurodegenerative disorders. The LPA receptor LPA1 appears to be involved in posthemorrhagic hydrocephalus. In a mouse model, LPA injection into the ventricle killed ependymal cells and produced hydrocephalus, but this was partially reduced in Lpar1 null mutant mice as well as with an LPA1 antagonist [110]. In glaucoma, the receptors LPA1 and LPA2 are upregulated in a rat model of elevated ocular pressure, and an LPA receptor agonist reduced histological damage and improved retinal electrophysiology [111]. In a different oxygen-induced retinopathy model in rats, an shRNA against Lpar1 was neuroprotective for RGC loss [112].

    Thus, in a variety of conditions, activation of LPA receptors appears to be pathological, while inhibition of LPA receptors appears neuroprotective.

    Interacting receptors: the PRGs

    There is also the possibility of interacting receptors that could modulate responses. For LPA receptors, one of the most intriguing possibilities is a family of receptors called the plasticity related genes (PRGs), also known as phospholipid phosphatase-related proteins (PLPPRs), which are a subfamily of the lipid phosphatase/phosphotransferase family of proteins (for review, see [113]). These are transmembrane proteins that are suggested to be receptors. Interestingly, although they are related to lipid phosphatases, they seem to have little or no lipid phosphatase activity.

    The first PRG, PRG-1 (also known as PRG1, PLPPR4), was identified as a gene that was upregulated during hippocampal development as well as after lesion [114]. Interestingly, PRG-1 overexpression in neuroblastoma cells counteracted the neurite retraction activity of LPA [114], suggesting a possible interaction. In addition, other evidence showed that another family member, PRG-3, could enhance axonal outgrowth in a variety of systems [115, 116]. Interaction with LPA receptors was demonstrated in a prg-1 null mutant mouse. Deletion of prg-1 led to epileptic seizures in the mouse (with larger synaptic currents and higher mEPSC frequencies), but simultaneous deletion of Lpar2 rescued this phenotype, suggesting an interaction [117]. However, the interaction may not be direct, as PRG-1 was localized postsynaptically and LPA2 was presynaptic. In other studies, another PRG, PRG-2, was found to be important for thalamocortical axon guidance to the barrel cortex. In this case, deletion of prg-2 resulted in misrouted thalamocortical axons, and this phenotype could be rescued by inhibiting autotaxin, the enzyme responsible for LPA biosynthesis [118].

    Furthermore, an interaction between LPA2 and PRG-1 has been seen in stroke. First, LPA levels increase in cerebrospinal fluid of human stroke patients. In a tMCAO mouse model of stroke, inhibition of autotaxin (which produces LPA) reduces LPA levels and improves behavioral outcomes with reduced lesion volume. Interestingly, a loss of function prg-1 mutation (R346T) resulted in increased lesion volume and worse behavioral outcomes after tMCAO [119]. An interaction with LPA2 was shown because deletion of Lpar2 along with the prg-1 mutation reversed the prg-1 mutant phenotype, resulting in lesion after tMCAO being similar to wild type injury after tMCAO. Thus, again, the Lpar2 mutation rescued the defects caused by a prg-1 mutation.

    Several studies have shown a role for PRGs to be neuroprotective and counteract the inhibitory activity of LPA and LPA receptors on axonal growth. PRG-2 promoted a growth state in neurons by binding and inhibiting PTEN (phosphatase and tensin homolog) [120]. PRG-3 is strongly expressed after spinal cord injury, and PRG-3 overexpression in cortical neurons induced neurite outgrowth and overcame inhibitory LPA treatment [121, 122]. The PRG family member PRG-5 induced filopodia when overexpressed, and its overexpression attenuated LPA-induced neurite retraction [123].

    A variety of experiments suggest an interaction of PRGs with LPA signaling through LPA receptors. In most cases, these receptors have not been identified, although an interaction between LPA2 and PRG-1 has been demonstrated genetically. Furthermore, whether there is direct interaction or not has yet to be established, as all the evidence so far shows only a functional interaction. Nonetheless, the potential interaction of PRGs with LPA receptors is intriguing, and much work remains to be done.

    Conclusions

    There has now accumulated substantial evidence that lysophospholipid GPCRs are involved in neurodegeneration and neuroprotection. This is especially true for S1P receptors due to the S1P receptor agonist, or functional antagonist, fingolimod, which has been clinically demonstrated to treat MS. The clinical success of fingolimod has led to the development of more specific S1P receptor pharmacological compounds as well as the investigation of S1P receptors in a variety of neurodegenerative conditions. There has been less investigation of LPA receptors in neurodegeneration, as there are fewer and less well-characterized pharmacological compounds as tools. Nonetheless, there is accumulating evidence that they, too, are involved in neuroprotection. As more LPA receptor compounds are developed and disease models examined, our understanding of the role of LPA receptors is likely to increase.

    Furthermore, there exists the intriguing possibility of interactions between certain GPCRs and other receptors, including other GPCRs. There is genetic evidence of a functional interaction of PRGs with LPA receptors, although there may not be direct physical interaction. A recent report suggests a direct physical interaction between LPA1 and the GPCR C-X-C motif chemokine receptor 4 (CXCR4) [124]. Interestingly, although coexpression of LPA1 and CXCR4 did not affect LPA signaling, signaling by the ligand C-X-C motif chemokine 12 (CXCL12) through CXCR4 is reduced by LPA treatment, suggesting a functional interaction [124]. Thus, an important direction for the future is to examine this concept of GPCR heterodimerization that could influence receptor signaling.

    Abbreviations

    ALS:

    amyotrophic lateral sclerosis

    BDNF:

    brain derived neurotrophic factor

    CNS:

    central nervous system

    CXCR4:

    C-X-C motif chemokine receptor 4

    EAE:

    experimental autoimmune encephalomyelitis

    ERK:

    extracellular signal-regulated kinase

    GPCR:

    G protein-coupled receptor

    LPA:

    lysophosphatidic acid

    MS:

    multiple sclerosis

    PRGs:

    plasticity related genes

    RR-MS:

    relapsing-remitting multiple sclerosis

    S1P:

    sphingosine-1-phosphate

    shRNA:

    short hairpin RNA

    tMCAO:

    transient middle cerebral artery occlusion

    Declarations

    Acknowledgments

    I would like to thank Dr. Daniel Stovall, Layla Herndon and Mallika Singh for critical reading of the manuscript.

    Author contributions

    EB: Writing—original draft, Writing—review & editing.

    Conflicts of interest

    The author declares no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    E.B. was supported by the National Institute of General Medical Sciences of the National Institutes of Health [P20GM103499]. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2024.

    References

    Hauser AS, Attwood MM, Rask-Andersen M, Schiöth HB, Gloriam DE. Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov. 2017;16:82942. [DOI] [PubMed] [PMC]
    Kihara Y, Maceyka M, Spiegel S, Chun J. Lysophospholipid receptor nomenclature review: IUPHAR Review 8. Br J Pharmacol. 2014;171:357594. [DOI] [PubMed] [PMC]
    Lucaciu A, Brunkhorst R, Pfeilschifter JM, Pfeilschifter W, Subburayalu J. The S1P-S1PR Axis in Neurological Disorders-Insights into Current and Future Therapeutic Perspectives. Cells. 2020;9:1515. [DOI] [PubMed] [PMC]
    Yanagida K, Masago K, Nakanishi H, Kihara Y, Hamano F, Tajima Y, et al. Identification and characterization of a novel lysophosphatidic acid receptor, p2y5/LPA6. J Biol Chem. 2009;284:1773141. [DOI] [PubMed] [PMC]
    Yanagida K, Shimizu T. Lysophosphatidic acid, a simple phospholipid with myriad functions. Pharmacol Ther. 2023;246:108421. [DOI] [PubMed]
    Birgbauer E. Lysophosphatidic Acid Signalling in Nervous System Development and Function. Neuromolecular Med. 2021;23:6885. [DOI] [PubMed]
    Xu Y. Targeting Lysophosphatidic Acid in Cancer: The Issues in Moving from Bench to Bedside. Cancers (Basel). 2019;11:1523. [DOI] [PubMed] [PMC]
    Meng H, Lee VM. Differential expression of sphingosine-1-phosphate receptors 1-5 in the developing nervous system. Dev Dyn. 2009;238:487500. [DOI] [PubMed]
    Choi JW, Chun J. Lysophospholipids and their receptors in the central nervous system. Biochim Biophys Acta. 2013;1831:2032. [DOI] [PubMed] [PMC]
    Im DS, Heise CE, Ancellin N, O’Dowd BF, Shei GJ, Heavens RP, et al. Characterization of a novel sphingosine 1-phosphate receptor, Edg-8. J Biol Chem. 2000;275:142816. [DOI] [PubMed]
    Terai K, Soga T, Takahashi M, Kamohara M, Ohno K, Yatsugi S, et al. Edg-8 receptors are preferentially expressed in oligodendrocyte lineage cells of the rat CNS. Neuroscience. 2003;116:105362. [DOI] [PubMed]
    Jaillard C, Harrison S, Stankoff B, Aigrot MS, Calver AR, Duddy G, et al. Edg8/S1P5: an oligodendroglial receptor with dual function on process retraction and cell survival. J Neurosci. 2005;25:145969. [DOI] [PubMed] [PMC]
    Brinkmann V, Davis MD, Heise CE, Albert R, Cottens S, Hof R, et al. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J Biol Chem. 2002;277:214537. [DOI] [PubMed]
    Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science. 2002;296:3469. [DOI] [PubMed]
    Hale JJ, Lynch CL, Neway W, Mills SG, Hajdu R, Keohane CA, et al. A rational utilization of high-throughput screening affords selective, orally bioavailable 1-benzyl-3-carboxyazetidine sphingosine-1-phosphate-1 receptor agonists. J Med Chem. 2004;47:66625. [DOI] [PubMed]
    Jo E, Sanna MG, Gonzalez-Cabrera PJ, Thangada S, Tigyi G, Osborne DA, et al. S1P1-selective in vivo-active agonists from high-throughput screening: off-the-shelf chemical probes of receptor interactions, signaling, and fate. Chem Biol. 2005;12:70315. [DOI] [PubMed]
    Sanna MG, Wang S, Gonzalez-Cabrera PJ, Don A, Marsolais D, Matheu MP, et al. Enhancement of capillary leakage and restoration of lymphocyte egress by a chiral S1P1 antagonist in vivo. Nat Chem Biol. 2006;2:43441. [DOI] [PubMed]
    Ohta H, Sato K, Murata N, Damirin A, Malchinkhuu E, Kon J, et al. Ki16425, a subtype-selective antagonist for EDG-family lysophosphatidic acid receptors. Mol Pharmacol. 2003;64:9941005. [DOI] [PubMed]
    Swaney JS, Chapman C, Correa LD, Stebbins KJ, Broadhead AR, Bain G, et al. Pharmacokinetic and pharmacodynamic characterization of an oral lysophosphatidic acid type 1 receptor-selective antagonist. J Pharmacol Exp Ther. 2011;336:693700. [DOI] [PubMed]
    Kozian DH, Evers A, Florian P, Wonerow P, Joho S, Nazare M. Selective non-lipid modulator of LPA5 activity in human platelets. Bioorg Med Chem Lett. 2012;22:523943. [DOI] [PubMed]
    Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, et al. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov. 2010;9:88397. [DOI] [PubMed]
    Chun J, Hartung H. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clin Neuropharmacol. 2010;33:91101. [DOI] [PubMed] [PMC]
    Kappos L, Radue E, O’Connor P, Polman C, Hohlfeld R, Calabresi P, et al.; FREEDOMS Study Group. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362:387401. [DOI] [PubMed]
    O’Sullivan S, Dev KK. Sphingosine-1-phosphate receptor therapies: Advances in clinical trials for CNS-related diseases. Neuropharmacology. 2017;113:597607. [DOI] [PubMed]
    Fujita T, Inoue K, Yamamoto S, Ikumoto T, Sasaki S, Toyama R, et al. Fungal metabolites. Part 11. A potent immunosuppressive activity found in Isaria sinclairii metabolite. J Antibiot (Tokyo). 1994;47:20815. [DOI] [PubMed]
    Adachi K, Kohara T, Nakao N, Arita M, Chiba K, Mishina T, et al. Design, synthesis, and structure-activity relationships of 2-substituted-2-amino-1,3-propanediols: Discovery of a novel immunosuppressant, FTY720. Bioorg Med Chem Lett. 1995;5:8536. [DOI]
    Suzuki S, Enosawa S, Kakefuda T, Shinomiya T, Amari M, Naoe S, et al. A novel immunosuppressant, FTY720, with a unique mechanism of action, induces long-term graft acceptance in rat and dog allotransplantation. Transplantation. 1996;61:2005. [DOI] [PubMed]
    Webb M, Tham C, Lin F, Lariosa-Willingham K, Yu N, Hale J, et al. Sphingosine 1-phosphate receptor agonists attenuate relapsing-remitting experimental autoimmune encephalitis in SJL mice. J Neuroimmunol. 2004;153:10821. [DOI] [PubMed]
    Foster CA, Howard LM, Schweitzer A, Persohn E, Hiestand PC, Balatoni B, et al. Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis. J Pharmacol Exp Ther. 2007;323:46975. [DOI] [PubMed]
    Balatoni B, Storch MK, Swoboda E, Schönborn V, Koziel A, Lambrou GN, et al. FTY720 sustains and restores neuronal function in the DA rat model of MOG-induced experimental autoimmune encephalomyelitis. Brain Res Bull. 2007;74:30716. [DOI] [PubMed]
    Choi JW, Gardell SE, Herr DR, Rivera R, Lee C, Noguchi K, et al. FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proc Natl Acad Sci U S A. 2011;108:7516. [DOI] [PubMed] [PMC]
    Ghasemi R, Dargahi L, Ahmadiani A. Integrated sphingosine-1 phosphate signaling in the central nervous system: From physiological equilibrium to pathological damage. Pharmacol Res. 2016;104:15664. [DOI] [PubMed]
    Gaire BP, Choi JW. Sphingosine 1-Phosphate Receptors in Cerebral Ischemia. Neuromolecular Med. 2021;23:21123. [DOI] [PubMed]
    Kimura A, Ohmori T, Kashiwakura Y, Ohkawa R, Madoiwa S, Mimuro J, et al. Antagonism of sphingosine 1-phosphate receptor-2 enhances migration of neural progenitor cells toward an area of brain. Stroke. 2008;39:34117. [DOI] [PubMed]
    Zhao P, Yang X, Yang L, Li M, Wood K, Liu Q, et al. Neuroprotective effects of fingolimod in mouse models of Parkinson’s disease. FASEB J. 2017;31:1729. [DOI] [PubMed]
    Rajan S, Sood A, Jain R, Kamatham PT, Khatri DK. Fingolimod exerts neuroprotection by regulating S1PR1 mediated BNIP3-PINK1-Parkin dependent mitophagy in rotenone induced mouse model of Parkinson’s disease. Neurosci Lett. 2024;820:137596. [DOI] [PubMed]
    Pyszko J, Strosznajder JB. Sphingosine kinase 1 and sphingosine-1-phosphate in oxidative stress evoked by 1-methyl-4-phenylpyridinium (MPP+) in human dopaminergic neuronal cells. Mol Neurobiol. 2014;50:3848. [DOI] [PubMed]
    Pardo AD, Amico E, Favellato M, Castrataro R, Fucile S, Squitieri F, et al. FTY720 (fingolimod) is a neuroprotective and disease-modifying agent in cellular and mouse models of Huntington disease. Hum Mol Genet. 2014;23:225165. [DOI] [PubMed]
    Potenza RL, Simone RD, Armida M, Mazziotti V, Pèzzola A, Popoli P, et al. Fingolimod: A Disease-Modifier Drug in a Mouse Model of Amyotrophic Lateral Sclerosis. Neurotherapeutics. 2016;13:91827. [DOI] [PubMed] [PMC]
    Deogracias R, Yazdani M, Dekkers MPJ, Guy J, Ionescu MCS, Vogt KE, et al. Fingolimod, a sphingosine-1 phosphate receptor modulator, increases BDNF levels and improves symptoms of a mouse model of Rett syndrome. Proc Natl Acad Sci U S A. 2012;109:142305. [DOI] [PubMed] [PMC]
    Kimura A, Ohmori T, Ohkawa R, Madoiwa S, Mimuro J, Murakami T, et al. Essential roles of sphingosine 1-phosphate/S1P1 receptor axis in the migration of neural stem cells toward a site of spinal cord injury. Stem Cells. 2007;25:11524. [DOI] [PubMed]
    Norimatsu Y, Ohmori T, Kimura A, Madoiwa S, Mimuro J, Seichi A, et al. FTY720 improves functional recovery after spinal cord injury by primarily nonimmunomodulatory mechanisms. Am J Pathol. 2012;180:162535. [DOI] [PubMed]
    Oizumi H, Sugimura Y, Totsune T, Kawasaki I, Ohshiro S, Baba T, et al. Plasma sphingolipid abnormalities in neurodegenerative diseases. PLoS One. 2022;17:e0279315. [DOI] [PubMed] [PMC]
    Asle-Rousta M, Kolahdooz Z, Oryan S, Ahmadiani A, Dargahi L. FTY720 (fingolimod) attenuates beta-amyloid peptide (Aβ42)-induced impairment of spatial learning and memory in rats. J Mol Neurosci. 2013;50:52432. [DOI] [PubMed]
    Hemmati F, Dargahi L, Nasoohi S, Omidbakhsh R, Mohamed Z, Chik Z, et al. Neurorestorative effect of FTY720 in a rat model of Alzheimer’s disease: comparison with memantine. Behav Brain Res. 2013;252:41521. [DOI] [PubMed]
    Fukumoto K, Mizoguchi H, Takeuchi H, Horiuchi H, Kawanokuchi J, Jin S, et al. Fingolimod increases brain-derived neurotrophic factor levels and ameliorates amyloid β-induced memory impairment. Behav Brain Res. 2014;268:8893. [DOI] [PubMed]
    Doi Y, Takeuchi H, Horiuchi H, Hanyu T, Kawanokuchi J, Jin S, et al. Fingolimod phosphate attenuates oligomeric amyloid β-induced neurotoxicity via increased brain-derived neurotrophic factor expression in neurons. PLoS One. 2013;8:e61988. [DOI] [PubMed] [PMC]
    Giguère FS, Essis SA, Chagniel L, Germain M, Cyr M, Massicotte G. The sphingosine-1-phosphate receptor 1 agonist SEW2871 reduces Tau-Ser262 phosphorylation in rat hippocampal slices. Brain Res. 2017;1658:519. [DOI] [PubMed]
    Wu Y, Mizugishi K, Bektas M, Sandhoff R, Proia RL. Sphingosine kinase 1/S1P receptor signaling axis controls glial proliferation in mice with Sandhoff disease. Hum Mol Genet. 2008;17:225764. [DOI] [PubMed] [PMC]
    Nakamura N, Honjo M, Yamagishi R, Kurano M, Yatomi Y, Watanabe S, et al. Neuroprotective role of sphingolipid rheostat in excitotoxic retinal ganglion cell death. Exp Eye Res. 2021;208:108623. [DOI] [PubMed]
    Edsall LC, Pirianov GG, Spiegel S. Involvement of sphingosine 1-phosphate in nerve growth factor-mediated neuronal survival and differentiation. J Neurosci. 1997;17:695260. [DOI] [PubMed] [PMC]
    Shinpo K, Kikuchi S, Moriwaka F, Tashiro K. Protective effects of the TNF-ceramide pathway against glutamate neurotoxicity on cultured mesencephalic neurons. Brain Res. 1999;819:1703. [DOI] [PubMed]
    Dusaban SS, Chun J, Rosen H, Purcell NH, Brown JH. Sphingosine 1-phosphate receptor 3 and RhoA signaling mediate inflammatory gene expression in astrocytes. J Neuroinflammation. 2017;14:111. [DOI] [PubMed] [PMC]
    Yung YC, Stoddard NC, Mirendil H, Chun J. Lysophosphatidic Acid signaling in the nervous system. Neuron. 2015;85:66982. [DOI] [PubMed] [PMC]
    Inoue M, Rashid MH, Fujita R, Contos JJA, Chun J, Ueda H. Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling. Nat Med. 2004;10:7128. [DOI] [PubMed]
    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. 2018;5:100020. [DOI] [PubMed] [PMC]
    Lin M, Rivera RR, Chun J. Targeted deletion of LPA5 identifies novel roles for lysophosphatidic acid signaling in development of neuropathic pain. J Biol Chem. 2012;287:1760817. [DOI] [PubMed] [PMC]
    Roza C, Campos-Sandoval JA, Gómez-García MC, Peñalver A, Márquez J. Lysophosphatidic Acid and Glutamatergic Transmission. Front Mol Neurosci. 2019;12:138. [DOI] [PubMed] [PMC]
    Ohuchi H, Hamada A, Matsuda H, Takagi A, Tanaka M, Aoki J, et al. Expression patterns of the lysophospholipid receptor genes during mouse early development. Dev Dyn. 2008;237:328094. [DOI] [PubMed]
    Suckau O, Gross I, Schrötter S, Yang F, Luo J, Wree A, et al. LPA1, LPA2, LPA4, and LPA6 receptor expression during mouse brain development. Dev Dyn. 2019;248:37595. [DOI] [PubMed] [PMC]
    Choi JW, Herr DR, Noguchi K, Yung YC, Lee C, Mutoh T, et al. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol. 2010;50:15786. [DOI] [PubMed]
    Yung YC, Stoddard NC, Chun J. LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res. 2014;55:1192214. [DOI] [PubMed] [PMC]
    Herr DR, Chew WS, Satish RL, Ong W. Pleotropic Roles of Autotaxin in the Nervous System Present Opportunities for the Development of Novel Therapeutics for Neurological Diseases. Mol Neurobiol. 2020;57:37292. [DOI] [PubMed]
    Benesch MGK, MacIntyre ITK, McMullen TPW, Brindley DN. Coming of Age for Autotaxin and Lysophosphatidate Signaling: Clinical Applications for Preventing, Detecting and Targeting Tumor-Promoting Inflammation. Cancers (Basel). 2018;10:73. [DOI] [PubMed] [PMC]
    Tigyi GJ, Yue J, Norman DD, Szabo E, Balogh A, Balazs L, et al. Regulation of tumor cell - Microenvironment interaction by the autotaxin-lysophosphatidic acid receptor axis. Adv Biol Regul. 2019;71:18393. [DOI] [PubMed] [PMC]
    Lee SC, Dacheux MA, Norman DD, Balázs L, Torres RM, Augelli-Szafran CE, et al. Regulation of Tumor Immunity by Lysophosphatidic Acid. Cancers (Basel). 2020;12:1202. [DOI] [PubMed] [PMC]
    Archbold JK, Martin JL, Sweet MJ. Towards selective lysophospholipid GPCR modulators. Trends Pharmacol Sci. 2014;35:21926. [DOI] [PubMed]
    Kingsbury MA, Rehen SK, Contos JJ, Higgins CM, Chun J. Non-proliferative effects of lysophosphatidic acid enhance cortical growth and folding. Nat Neurosci. 2003;6:12929. [DOI] [PubMed]
    Estivill-Torrús G, Llebrez-Zayas P, Matas-Rico E, Santín L, Pedraza C, Del Diego I, et al. Absence of LPA1 signaling results in defective cortical development. Cereb Cortex. 2008;18:93850. [DOI] [PubMed]
    Matas-Rico E, García-Diaz B, Llebrez-Zayas P, López-Barroso D, Santín L, Pedraza C, et al. Deletion of lysophosphatidic acid receptor LPA1 reduces neurogenesis in the mouse dentate gyrus. Mol Cell Neurosci. 2008;39:34255. [DOI] [PubMed] [PMC]
    Santin LJ, Bilbao A, Pedraza C, Matas-Rico E, López-Barroso D, Castilla-Ortega E, et al. Behavioral phenotype of maLPA1-null mice: increased anxiety-like behavior and spatial memory deficits. Genes Brain Behav. 2009;8:77284. [DOI] [PubMed] [PMC]
    Castilla-Ortega E, Sánchez-López J, Hoyo-Becerra C, Matas-Rico E, Zambrana-Infantes E, Chun J, et al. Exploratory, anxiety and spatial memory impairments are dissociated in mice lacking the LPA1 receptor. Neurobiol Learn Mem. 2010;94:7382. [DOI] [PubMed] [PMC]
    Jalink K, Eichholtz T, Postma FR, van Corven EJ, Moolenaar WH. Lysophosphatidic acid induces neuronal shape changes via a novel, receptor-mediated signaling pathway: similarity to thrombin action. Cell Growth Differ. 1993;4:24755. [PubMed]
    Tigyi G, Fischer DJ, Sebök A, Marshall F, Dyer DL, Miledi R. Lysophosphatidic acid-induced neurite retraction in PC12 cells: neurite-protective effects of cyclic AMP signaling. J Neurochem. 1996;66:54958. [DOI] [PubMed]
    Saito S. Effects of lysophosphatidic acid on primary cultured chick neurons. Neurosci Lett. 1997;229:736. [DOI] [PubMed]
    Campbell DS, Holt CE. Chemotropic responses of retinal growth cones mediated by rapid local protein synthesis and degradation. Neuron. 2001;32:101326. [DOI] [PubMed]
    Sayas CL, Avila J, Wandosell F. Glycogen synthase kinase-3 is activated in neuronal cells by Gα12 and Gα13 by Rho-independent and Rho-dependent mechanisms. J Neurosci. 2002;22:686375. [DOI] [PubMed] [PMC]
    Birgbauer E, Chun J. Lysophospholipid receptors LPA1-3 are not required for the inhibitory effects of LPA on mouse retinal growth cones. Eye Brain. 2010;2:113. [DOI] [PubMed] [PMC]
    Fincher J, Whiteneck C, Birgbauer E. G-protein-coupled receptor cell signaling pathways mediating embryonic chick retinal growth cone collapse induced by lysophosphatidic acid and sphingosine-1-phosphate. Dev Neurosci. 2014;36:44353. [DOI] [PubMed] [PMC]
    Jalink K, van Corven EJ, Hengeveld T, Morii N, Narumiya S, Moolenaar WH. Inhibition of lysophosphatidate- and thrombin-induced neurite retraction and neuronal cell rounding by ADP ribosylation of the small GTP-binding protein Rho. J Cell Biol. 1994;126:80110. [DOI] [PubMed] [PMC]
    Tigyi G, Fischer DJ, Sebök A, Yang C, Dyer DL, Miledi R. Lysophosphatidic acid-induced neurite retraction in PC12 cells: control by phosphoinositide-Ca2+ signaling and Rho. J Neurochem. 1996;66:53748. [DOI] [PubMed]
    Kozma R, Sarner S, Ahmed S, Lim L. Rho family GTPases and neuronal growth cone remodelling: relationship between increased complexity induced by Cdc42Hs, Rac1, and acetylcholine and collapse induced by RhoA and lysophosphatidic acid. Mol Cell Biol. 1997;17:120111. [DOI] [PubMed] [PMC]
    Hirose M, Ishizaki T, Watanabe N, Uehata M, Kranenburg O, Moolenaar WH, et al. Molecular dissection of the Rho-associated protein kinase (p160ROCK)-regulated neurite remodeling in neuroblastoma N1E-115 cells. J Cell Biol. 1998;141:162536. [DOI] [PubMed] [PMC]
    Kranenburg O, Poland M, van Horck FP, Drechsel D, Hall A, Moolenaar WH. Activation of RhoA by lysophosphatidic acid and Gα12/13 subunits in neuronal cells: induction of neurite retraction. Mol Biol Cell. 1999;10:18517. [DOI] [PubMed] [PMC]
    Bito H, Furuyashiki T, Ishihara H, Shibasaki Y, Ohashi K, Mizuno K, et al. A critical role for a Rho-associated kinase, p160ROCK, in determining axon outgrowth in mammalian CNS neurons. Neuron. 2000;26:43141. [DOI] [PubMed]
    Yamazaki J, Katoh H, Negishi M. Lysophosphatidic acid and thrombin receptors require both Gα12 and Gα13 to regulate axonal morphology in hippocampal neurons. Biol Pharm Bull. 2008;31:221622. [DOI] [PubMed]
    Furuta D, Yamane M, Tsujiuchi T, Moriyama R, Fukushima N. Lysophosphatidic acid induces neurite branch formation through LPA3. Mol Cell Neurosci. 2012;50:2134. [DOI] [PubMed]
    Frugier T, Crombie D, Conquest A, Tjhong F, Taylor C, Kulkarni T, et al. Modulation of LPA receptor expression in the human brain following neurotrauma. Cell Mol Neurobiol. 2011;31:56977. [DOI] [PubMed]
    Crack PJ, Zhang M, Morganti-Kossmann MC, Morris AJ, Wojciak JM, Fleming JK, et al. Anti-lysophosphatidic acid antibodies improve traumatic brain injury outcomes. J Neuroinflammation. 2014;11:37. [DOI] [PubMed] [PMC]
    Goldshmit Y, Munro K, Leong SY, Pébay A, Turnley AM. LPA receptor expression in the central nervous system in health and following injury. Cell Tissue Res. 2010;341:2332. [DOI] [PubMed]
    Santos-Nogueira E, López-Serrano C, Hernández J, Lago N, Astudillo AM, Balsinde J, et al. Activation of Lysophosphatidic Acid Receptor Type 1 Contributes to Pathophysiology of Spinal Cord Injury. J Neurosci. 2015;35:1022435. [DOI] [PubMed] [PMC]
    López-Serrano C, Santos-Nogueira E, Francos-Quijorna I, Coll-Miró M, Chun J, López-Vales R. Lysophosphatidic acid receptor type 2 activation contributes to secondary damage after spinal cord injury in mice. Brain Behav Immun. 2019;76:25867. [DOI] [PubMed] [PMC]
    Fink KL, López-Giráldez F, Kim I, Strittmatter SM, Cafferty WBJ. Identification of Intrinsic Axon Growth Modulators for Intact CNS Neurons after Injury. Cell Rep. 2017;18:2687701. [DOI] [PubMed] [PMC]
    Li Z, Yu Z, Wang D, Ju W, Zhan X, Wu Q, et al. Influence of acetylsalicylate on plasma lysophosphatidic acid level in patients with ischemic cerebral vascular diseases. Neurol Res. 2008;30:3669. [DOI] [PubMed]
    Li Z, Yu Z, Yu Y, Ju W, Wang D, Zhan X, et al. Lysophosphatidic acid level and the incidence of silent brain infarction in patients with nonvalvular atrial fibrillation. Int J Mol Sci. 2010;11:398898. [DOI] [PubMed] [PMC]
    Wang C, Zhang J, Tang J, Li Y, Gu Y, Yu Y, et al. Lysophosphatidic acid induces neuronal cell death via activation of asparagine endopeptidase in cerebral ischemia-reperfusion injury. Exp Neurol. 2018;306:19. [DOI] [PubMed]
    Zeng X, Luo Z, Wu J, Zhou J, Shan Y, Zhu Y, et al. Cerebral Ischemia-Reperfusion Injury: Lysophosphatidic Acid Mediates Inflammation by Decreasing the Expression of Liver X Receptor. J Mol Neurosci. 2020;70:137684. [DOI] [PubMed]
    Chi OZ, Mellender SJ, Kiss GK, Chiricolo A, Liu X, Patel N, et al. Lysophosphatidic acid increased infarct size in the early stage of cerebral ischemia-reperfusion with increased BBB permeability. J Stroke Cerebrovasc Dis. 2020;29:105029. [DOI] [PubMed]
    Weiss HR, Mellender SJ, Kiss GK, Chiricolo A, Liu X, Chi OZ. Lysophosphatidic Acid Reduces Microregional Oxygen Supply/Consumption Balance after Cerebral Ischemia-Reperfusion. J Vasc Res. 2020;57:17884. [DOI] [PubMed]
    Gaire BP, Sapkota A, Song M, Choi JW. Lysophosphatidic acid receptor 1 (LPA1) plays critical roles in microglial activation and brain damage after transient focal cerebral ischemia. J Neuroinflammation. 2019;16:170. [DOI] [PubMed] [PMC]
    Sapkota A, Lee C, Park SJ, Choi JW. Lysophosphatidic Acid Receptor 5 Plays a Pathogenic Role in Brain Damage after Focal Cerebral Ischemia by Modulating Neuroinflammatory Responses. Cells. 2020;9:1446. [DOI] [PubMed] [PMC]
    Plastira I, Bernhart E, Goeritzer M, Reicher H, Kumble VB, Kogelnik N, et al. 1-Oleyl-lysophosphatidic acid (LPA) promotes polarization of BV-2 and primary murine microglia towards an M1-like phenotype. J Neuroinflammation. 2016;13:205. [DOI] [PubMed] [PMC]
    Plastira I, Bernhart E, Goeritzer M, DeVaney T, Reicher H, Hammer A, et al. Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype. J Neuroinflammation. 2017;14:253. [DOI] [PubMed] [PMC]
    Hirata T, Cui YJ, Funakoshi T, Mizukami Y, Ishikawa Y, Shibasaki F, et al. The temporal profile of genomic responses and protein synthesis in ischemic tolerance of the rat brain induced by repeated hyperbaric oxygen. Brain Res. 2007;1130:21422. [DOI] [PubMed]
    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. [DOI] [PubMed] [PMC]
    Hao Y, Guo M, Feng Y, Dong Q, Cui M. Lysophospholipids and Their G-Coupled Protein Signaling in Alzheimer’s Disease: From Physiological Performance to Pathological Impairment. Front Mol Neurosci. 2020;13:58. [DOI] [PubMed] [PMC]
    Sayas CL, Moreno-Flores MT, Avila J, Wandosell F. The neurite retraction induced by lysophosphatidic acid increases Alzheimer’s disease-like Tau phosphorylation. J Biol Chem. 1999;274:3704652. [DOI] [PubMed]
    de San Román EG, Llorente-Ovejero A, Martínez-Gardeazabal J, Moreno-Rodríguez M, Giménez-Llort L, Manuel I, et al. Modulation of Neurolipid Signaling and Specific Lipid Species in the Triple Transgenic Mouse Model of Alzheimer’s Disease. Int J Mol Sci. 2021;22:12256. [DOI] [PubMed] [PMC]
    Puigdomenech-Poch M, Martínez-Muriana A, Andrés-Benito P, Ferrer I, Chun J, López-Vales R. Dual Role of Lysophosphatidic Acid Receptor 2 (LPA2) in Amyotrophic Lateral Sclerosis. Front Cell Neurosci. 2021;15:600872. [DOI] [PubMed] [PMC]
    Lummis NC, Sánchez-Pavón P, Kennedy G, Frantz AJ, Kihara Y, Blaho VA, et al. LPA1/3 overactivation induces neonatal posthemorrhagic hydrocephalus through ependymal loss and ciliary dysfunction. Sci Adv. 2019;5:eaax2011. [DOI] [PubMed] [PMC]
    Savitz SI, Dhallu MS, Malhotra S, Mammis A, Ocava LC, Rosenbaum PS, et al. EDG receptors as a potential therapeutic target in retinal ischemia-reperfusion injury. Brain Res. 2006;1118:16875. [DOI] [PubMed]
    Yang C, Lafleur J, Mwaikambo BR, Zhu T, Gagnon C, Chemtob S, et al. The role of lysophosphatidic acid receptor (LPA1) in the oxygen-induced retinal ganglion cell degeneration. Invest Ophthalmol Vis Sci. 2009;50:12908. [DOI] [PubMed]
    Fuchs J, Bareesel S, Kroon C, Polyzou A, Eickholt BJ, Leondaritis G. Plasma membrane phospholipid phosphatase-related proteins as pleiotropic regulators of neuron growth and excitability. Front Mol Neurosci. 2022;15:984655. [DOI] [PubMed] [PMC]
    Bräuer AU, Savaskan NE, Kühn H, Prehn S, Ninnemann O, Nitsch R. A new phospholipid phosphatase, PRG-1, is involved in axon growth and regenerative sprouting. Nat Neurosci. 2003;6:5728. [DOI] [PubMed]
    Sigal YJ, Quintero OA, Cheney RE, Morris AJ. Cdc42 and ARP2/3-independent regulation of filopodia by an integral membrane lipid-phosphatase-related protein. J Cell Sci. 2007;120:34052. [DOI] [PubMed]
    Velmans T, Battefeld A, Geist B, Farrés AS, Strauss U, Bräuer AU. Plasticity-related gene 3 promotes neurite shaft protrusion. BMC Neurosci. 2013;14:36. [DOI] [PubMed] [PMC]
    Trimbuch T, Beed P, Vogt J, Schuchmann S, Maier N, Kintscher M, et al. Synaptic PRG-1 modulates excitatory transmission via lipid phosphate-mediated signaling. Cell. 2009;138:122235. [DOI] [PubMed] [PMC]
    Cheng J, Sahani S, Hausrat TJ, Yang J, Ji H, Schmarowski N, et al. Precise Somatotopic Thalamocortical Axon Guidance Depends on LPA-Mediated PRG-2/Radixin Signaling. Neuron. 2016;92:12642. [DOI] [PubMed] [PMC]
    Bitar L, Uphaus T, Thalman C, Muthuraman M, Gyr L, Ji H, et al. Inhibition of the enzyme autotaxin reduces cortical excitability and ameliorates the outcome in stroke. Sci Transl Med. 2022;14:eabk0135. [DOI] [PubMed]
    Brosig A, Fuchs J, Ipek F, Kroon C, Schrötter S, Vadhvani M, et al. The Axonal Membrane Protein PRG2 Inhibits PTEN and Directs Growth to Branches. Cell Rep. 2019;29:202840.e8. [DOI] [PubMed] [PMC]
    Broggini T, Schnell L, Ghoochani A, Mateos JM, Buchfelder M, Wiendieck K, et al. Plasticity Related Gene 3 (PRG3) overcomes myelin-associated growth inhibition and promotes functional recovery after spinal cord injury. Aging (Albany NY). 2016;8:246387. [DOI] [PubMed] [PMC]
    Iweka CA, Hussein RK, Yu P, Katagiri Y, Geller HM. The lipid phosphatase-like protein PLPPR1 associates with RhoGDI1 to modulate RhoA activation in response to axon growth inhibitory molecules. J Neurochem. 2021;157:494507. [DOI] [PubMed] [PMC]
    Broggini T, Nitsch R, Savaskan NE. Plasticity-related gene 5 (PRG5) induces filopodia and neurite growth and impedes lysophosphatidic acid- and nogo-A-mediated axonal retraction. Mol Biol Cell. 2010;21:52137. [DOI] [PubMed] [PMC]
    Hong JM, Lee J, Seen D, Jeong J, Huh W. LPA1-mediated inhibition of CXCR4 attenuates CXCL12-induced signaling and cell migration. Cell Commun Signal. 2023;21:257. [DOI] [PubMed] [PMC]