Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2013 May 20;110(23):9529–9534. doi: 10.1073/pnas.1219004110

GPR37 and GPR37L1 are receptors for the neuroprotective and glioprotective factors prosaptide and prosaposin

Rebecca C Meyer 1, Michelle M Giddens 1, Stacy A Schaefer 1, Randy A Hall 1,1
PMCID: PMC3677493  PMID: 23690594

Abstract

GPR37 (also known as Pael-R) and GPR37L1 are orphan G protein-coupled receptors that are almost exclusively expressed in the nervous system. We screened these receptors for potential activation by various orphan neuropeptides, and these screens yielded a single positive hit: prosaptide, which promoted the endocytosis of GPR37 and GPR37L1, bound to both receptors and activated signaling in a GPR37- and GPR37L1-dependent manner. Prosaptide stimulation of cells transfected with GPR37 or GPR37L1 induced the phosphorylation of ERK in a pertussis toxin-sensitive manner, stimulated 35S-GTPγS binding, and promoted the inhibition of forskolin-stimulated cAMP production. Because prosaptide is the active fragment of the secreted neuroprotective and glioprotective factor prosaposin (also known as sulfated glycoprotein-1), we purified full-length prosaposin and found that it also stimulated GPR37 and GPR37L1 signaling. Moreover, both prosaptide and prosaposin were found to protect primary astrocytes against oxidative stress, with these protective effects being attenuated by siRNA-mediated knockdown of endogenous astrocytic GPR37 or GPR37L1. These data reveal that GPR37 and GPR37L1 are receptors for the neuroprotective and glioprotective factors prosaptide and prosaposin.

Keywords: deorphanization, GPCR, MAPK, neurodegeneration, peptide


Approximately half of all drugs in current clinical use act on G protein-coupled receptors (GPCRs) (1). GPCRs are outstanding drug targets for a variety of reasons, including their localization at the cell surface, their natural propensity for being activated by small molecules (neurotransmitters, hormones, sensory stimuli, etc.), and in many cases their discrete patterns of distribution in the body, which can allow for tissue-specific targeting of therapeutics. There are still more than 100 GPCRs with unknown ligands, and these so-called orphan receptors represent a large pool of potential targets for novel therapeutics (2). However, drug development efforts aimed at GPCRs are extremely difficult in the absence of a known ligand.

Two orphan GPCRs that have attracted significant interest over the past 15 years are GPR37 and GPR37L1, a pair of closely related receptors that exhibit more distant similarity to endothelin receptors and other peptide-activated GPCRs (39). GPR37 and GPR37L1 are found almost exclusively in the nervous system and are known to be expressed in both neurons and glia (3, 4, 610). GPR37 has been studied with particular intensity since it was identified as a substrate of the E3 ubiquitin ligase parkin, earning it the alternative name “parkin-associated endothelin-like receptor,” or “Pael-R” (9). Although parkin has many substrates (11), GPR37 may be one of the key substrates related to pathology in patients with parkin mutations, which are associated with autosomal recessive juvenile Parkinson disease (9, 12, 13). The connection between GPR37 and parkin has led to a focus on the dopaminergic system in GPR37 knockout mice, which exhibit a reduced dopaminergic tone and various subtle perturbations in dopaminergic signaling in the brain (1416). However, GPR37 and GPR37L1 are expressed in many different brain regions beyond just dopaminergic areas, so it seems probable that these receptors play a broader role in the regulation of neuronal and glial physiology beyond simply modulating dopaminergic neurotransmission.

Because GPR37 and GPR37L1 exhibit their strongest sequence similarity to the endothelin receptors and other GPCRs activated by peptides, it has been viewed as likely that these receptors must be peptide-activated. Indeed, it has been reported that GPR37 can bind to a neuropeptide known as “head activator” (HA) (17, 18), which is derived from the invertebrate Hydra. However, following a handful of reports three decades ago about the potential existence of an HA ortholog in mammalian brains (19, 20), no further evidence for an HA ortholog in vertebrates has come to light. Several groups have explored the possibility that GPR37 and/or GPR37L1 might be activated by endothelins or endothelin-related peptides, but all studies of this type have yielded negative results (4, 7). Thus, GPR37 and GPR37L1 remain orphan receptors. Here we present evidence that these receptors are activated by the neuroprotective and glioprotective factor prosaposin (also known as sulfated glycoprotein-1) as well as by prosaptide, a peptide derived from prosaposin. Both prosaposin and prosaptide have been reported in numerous studies over the past two decades to exert neuroprotective and glioprotective effects (2142) via the stimulation of G protein-mediated pathways (25, 28, 35, 37, 43), but the receptor(s) mediating these effects have not been identified. Our data reveal that GPR37 and GPR37L1 are sufficient for evoking prosaptide/prosaposin-mediated responses in HEK-293T cells and necessary for mediating endogenous responses to prosaptide/prosaposin in primary cortical astrocytes.

Results

Prosaptide Binds to and Induces Endocytosis of GPR37 and GPR37L1.

A major impediment to identifying the natural ligand for GPR37 has been the poor trafficking of the receptor to the plasma membrane upon transfection into most cell types (44). Because we demonstrated that interaction of GPR37 with the PDZ scaffold protein syntenin-1 greatly promotes trafficking of GPR37 to the plasma membrane in heterologous cells (44), we performed screens for potential GPR37 ligands using HEK-293T cells cotransfected with GPR37 and syntenin-1. In parallel studies, we also screened cells transfected with GPR37L1, which is trafficked to the plasma membrane in HEK-293T cells efficiently without syntenin-1 or other coexpressed chaperone proteins (44). The expression plasmids used in these studies confer Flag tags on the extracellular N termini of GPR37 and GPR37L1. As a screening assay, we examined internalization of the Flag tag from the cell surface in response to ligand stimulation using cell-surface luminometry. Because most GPCRs undergo significant endocytosis in response to agonist stimulation (45), ligand-induced endocytosis of a receptor is a strong indication that the ligand may be an agonist for the receptor.

In screens for potential ligand-induced endocytosis of GPR37 and GPR37L1, we examined five neuropeptides— secretoneurin (46), substance P 1–7 (47), cocaine- and amphetamine-regulated transcript (CART) (48, 49), prosaptide (25, 28, 29, 35, 37, 43), and l-prolyl-l-leucyl-glycinamide (PLG) (50)—which are all known to exert physiological effects via G protein-dependent pathways in cell types and/or areas of the brain that express GPR37 and GPR37L1. For four of these five peptides, we observed no effect on GPR37 or GPR37L1 surface expression following a 30-min peptide treatment (1 µM). However, treatment with one of the peptides, prosaptide, induced robust endocytosis of both GPR37 and GPR37L1 (Fig. 1 A and B). Prosaptide-induced internalization of GPR37L1 was further demonstrated in antibody feeding experiments in which surface-expressed receptors on live cells were labeled with an antibody against the N-terminal Flag tag before stimulation with prosaptide. Treatment with prosaptide (1 µM, 30 min) induced robust endocytosis of the antibody-labeled receptors (Fig. 1 C and D; internalized receptors are indicated by the green labeling). This prosaptide-induced internalization of GPR37L1 in the confocal microscopy experiments was quantified with regard to the ratio of internalized to cell surface receptor (Fig. 1E), thereby confirming the results of the cell surface luminometry studies using a second independent technique.

Fig. 1.

Fig. 1.

Prosaptide binds to and induces endocytosis of GPR37 and GPR37L1. (A and B) Five different peptides were examined for their abilities to induce internalization of Flag-tagged GPR37 (A) and GPR37L1 (B) in HEK-293T cells. Each peptide was added to the cells at 1 µM for 30 min. Only treatment with prosaptide induced significant receptor internalization (n = 3–5 for each peptide; all points done in triplicate, **P < 0.01, ***P < 0.001). (C and D) The ability of prosaptide to induce internalization of GPR37L1 in transfected COS-7 cells was visualized using confocal microscopy. Cell surface GPR37L1 is labeled in red, with green labeling indicating internalized GPR37L1. Representative images of GPR37L1 localization in vehicle-treated cells (C) and cells treated with 1 µM prosaptide (D) are shown. (E) Quantification of surface vs. internalized GPR37L1. COS-7 cells were analyzed for ratio of internalization by comparing the quantity of internalized receptor (green) to the total receptor found on the cell surface (red). In this analysis, 20 cells each from vehicle- vs. prosaptide-treated GPR37L1-transfected COS-7 cells were randomly selected in a blinded manner and analyzed to assess the extent of GPR37L1 internalization (***P < 0.001). (F) The ability of prosaptide to bind to different GPCRs was assessed using biotinylated prosaptide attached to streptavidin beads (+) or streptavidin beads alone (−). Flag-tagged GPR37 and GPR37L1, but not other Flag-tagged (α1A- and β1-adrenergic) or HA-tagged receptors (D1 dopamine and endothelin ETB), were pulled down with the biotinylated prosaptide adsorbed to the beads, indicating specific binding of the peptide to GPR37 and GPR37L1.

Following our observations of prosaptide-induced internalization of GPR37 and GPR37L1, we sought to determine whether prosaptide could bind to these receptors. Studies with the endothelin receptors, the most closely related receptors to GPR37/GPR37L1, have demonstrated that solubilized endothelin receptors can be efficiently pulled down by biotinylated endothelins coupled to streptavidin (51, 52). Thus, we obtained a biotinylated version of prosaptide and pursued a similar approach for studying prosaptide binding to GPR37 and GPR37L1. As shown in Fig. 1F, solubilized GPR37 and GPR37L1 were robustly pulled down by biotinylated prosaptide but not by control streptavidin beads. In contrast, a number of other Flag- or HA-tagged transfected GPCRs (α1A-adrenergic, β1-adrenergic, D1 dopamine, and endothelin ETB receptors) were not detectably pulled down by biotinylated prosaptide.

Prosaptide Stimulates G Protein-Mediated Signaling via GPR37 and GPR37L1.

Given that the above-described experiments revealed prosaptide could bind to GPR37 and GPR37L1 and induce the internalization of these receptors, we next turned our attention to examining whether prosaptide could activate GPR37 and GPR37L1 to induce intracellular signaling. The most frequently reported signaling pathway activated by prosaptide is the stimulation of ERK phosphorylation (26, 28, 5355). Thus, we examined the ability of prosaptide to induce ERK phosphorylation in HEK-293T cells transfected with either GPR37/syntenin-1 or GPR37L1. Transfection of the receptors into HEK-293T cells had no significant effect on basal levels of phospho-ERK or total ERK (Fig. S1), but treatment with 100 nM prosaptide induced a significant increase in pERK for both GPR37- and GPR37L1-transfected cells, with no effect observed on mock-transfected cells (Fig. 2 A and B). Dose–response studies revealed that prosaptide activates GPR37 with an apparent EC50 of 7 nM (Fig. 2C) and GPR37L1 with an EC50 of 5 nM (Fig. 2D). Because prosaptide has been reported to activate a pertussis toxin (PTX)-sensitive Gαi/o-coupled receptor (25, 28, 29, 35, 37, 43), we examined the effects of PTX pretreatment and found that it completely inhibited the ability of prosaptide to stimulate ERK phosphorylation in GPR37- or GPR37L1-transfected cells: pERK induced by prosaptide in the presence of PTX was significantly lower than prosaptide stimulated pERK without PTX (Figs. S2 and S3). These data indicate a Gαi/o-dependent mechanism of ERK activation for these receptors. We also found that prosaptide significantly inhibited forskolin-stimulated cAMP production in cells transfected with GPR37/syntenin-1 or GPR37L1 (Fig. 2E) and enhanced 35S-GTPγS binding to membranes derived from cells transfected with GPR37/syntenin-1/Gαi1 or GPR37L1/Gαi1 (Fig. 2F), consistent with the idea that prosaptide stimulation promotes coupling of Gαi/o to GPR37 and GPR37L1.

Fig. 2.

Fig. 2.

Prosaptide stimulates G protein-mediated signaling via GPR37 and GPR37L1. (A) HEK-293T cells transiently transfected with either empty vector (Mock), GPR37/syntenin-1 or GPR37L1 were treated for 10 min with 100 nM prosaptide and ERK phosphorylation was assessed. pERK, phosphorylated ERK; tERK, total ERK. (B) Quantification of changes in pERK levels in response to prosaptide treatment. All experiments determined prosaptide-induced ERK phosphorylation over vehicle treatment (n = 3; all points done in duplicate, ***P < 0.001). (C and D) Dose–response curves for prosaptide stimulation of HEK-293T cells transfected with either GPR37/syntenin-1 (C) or GPR37L1 (D) (n = 6). (E) Effects of prosaptide treatment (30 min, 100 nM) on cAMP levels in HEK-293T cells transfected with empty vector (Mock), GPR37/syntenin-1, or GPR37L1. All experiments determined prosaptide-induced cAMP inhibition compared with vehicle treatment (n = 5; all points done in triplicate, **P < 0.01). (F) Effects of prosaptide treatment on 35S-GTPγS binding to membranes derived from HEK-293T cells transfected with Gαi1 alone (Mock), GPR37/syntenin-1/Gαi1, or GPR37L1/Gαi1. All experiments determined prosaptide-induced 35S-GTPγS binding over vehicle treatment (n = 3; all points done in duplicate, **P < 0.01, *P < 0.05). Total counts for vehicle-treated samples averaged ∼6,000 cpm and did not vary significantly between transfection conditions.

Prosaptide Actions on Primary Astrocytes Are Mediated by GPR37 and GPR37L1.

All of the experiments described above were performed using cells transfected with GPR37 and GPR37L1 to provide evidence that transfection with these receptors is sufficient to confer physiological responses to prosaptide. To address the issue of whether GPR37 and/or GPR37L1 is necessary for endogenous prosaptide-induced signaling, we turned to primary cultures of cortical astrocytes, a cell type known to express mRNA for both GPR37 and GPR37L1 (10, 56). We confirmed that primary cortical astrocytes express GPR37 and GPR37L1 at the protein level via Western blotting using antibodies specific to each receptor (Fig. 3A) using antibodies that were determined in studies on transfected cells to be specific for each receptor (Figs. S4 and S5). These studies revealed that GPR37 is more abundantly expressed in cortical astrocytes relative to whole brain (per unit protein), whereas GPR37L1 is more enriched in whole brain relative to primary cortical astrocytes.

Fig. 3.

Fig. 3.

Prosaptide actions on primary astrocytes are mediated by GPR37 and GPR37L1. (A) The relative expression levels of GPR37 and GPR37L1 in primary cortical astrocyte lysates vs. whole-brain lysates were assessed via Western blot. (B) Prosaptide-induced ERK phosphorylation in primary cortical astrocytes was assessed under several conditions: no siRNA treatment, treatment with a scrambled siRNA, or treatment with siRNAs directed against GPR37 and/or GPR37L1. KD, knockdown. (C) Quantification of the studies illustrated in B (n = 3; all points done in duplicate, ***P < 0.001). (D) Prosaptide-mediated protection of cortical astrocytes from a hydrogen peroxide insult was assessed by measuring lactate dehydrogenase release. Labels are the same as in C (n = 3; all points done in triplicate, ***P < 0.001).

Treatment with prosaptide (100 nM) induced robust ERK phosphorylation in the primary cortical astrocyte cultures (Fig. 3 B and C). To determine whether this effect was mediated by GPR37 and/or GPR37L1, we knocked down expression of each receptor using a siRNA approach (Fig. S6). Treatment of the cells with either scrambled siRNA or siRNA knocking down GPR37L1 had no significant effect on prosaptide-mediated pERK stimulation. Conversely, knockdown of GPR37 or the joint knockdown of GPR37 and GPR37L1 together completely abolished the ability of prosaptide to induce ERK phosphorylation (Fig. 3 B and C). It is important to point out that G protein-mediated regulation of the ERK pathway was still intact following siRNA-mediated knockdown of GPR37 and GPR37L1, as assessed by the ability of endogenous lysophosphatidic acid receptors in the astrocytes to stimulate increases in pERK (Fig. S7), thereby suggesting that the siRNA treatments did not induce any nonspecific impairments in the ERK pathway due to off-target effects.

Because prosaptide has been reported to act as a neuroprotective and glioprotective factor (21, 2340, 42, 57), we also studied the ability of prosaptide to protect primary astrocytes against oxidative stress induced by hydrogen peroxide (H2O2). Cortical astrocytes were pretreated with either vehicle or 100 nM prosaptide for 10 min then treated with 500 µM H2O2 for 24 h and assessed for the release of lactate dehydrogenase into the media as an indicator of cell death. Prosaptide treatment significantly protected the cortical astrocytes from H2O2-induced death in cultures treated with either no siRNA or scrambled siRNA. However, in astrocytes in which either GPR37, GPR37L1, or both receptors had been knocked down, the ability of prosaptide to promote cell survival was significantly attenuated (Fig. 3D).

Full-Length Prosaposin Protects Primary Astrocytes via Activation of GPR37 and GPR37L1.

Prosaptide is a peptide fragment that mimics the trophic and protective actions of full-length prosaposin (23, 24, 26, 28, 32, 35, 36, 38, 53, 54), but it is widely presumed that full-length prosaposin is the endogenous ligand for any receptor that can be activated by prosaptide. Thus, we purified full-length prosaposin using a slightly modified version of a previously described protocol (58) and assessed whether full-length prosaposin could activate GPR37 and GPR37L1. Cells transfected with empty vector, GPR37/syntenin-1, or GPR37L1 were incubated with purified prosaposin (100 nM). Treatment with prosaposin induced ERK phosphorylation in the cells expressing GPR37/syntenin-1 or GPR37L1, but not in mock transfected cells (Fig. 4A), similar to the results obtained earlier in the prosaptide stimulation experiments. Full-length prosaposin also induced internalization of GPR37 and GPR37L1 (Fig. 4B), again mirroring our earlier findings with prosaptide.

Fig. 4.

Fig. 4.

Full-length prosaposin protects primary astrocytes via activation of GPR37 and GPR37L1. (A) Treatment of HEK-293T cells transiently transfected with empty vector (Mock), GPR37/syntenin-1, or GPR37L1 with prosaposin (100 nM, 10 min) resulted in enhanced ERK phosphorylation in only GPR37- or GPR37L1-transfected cells (n = 3; all points done in duplicate, ***P < 0.001). (B) Endocytosis of Flag-GPR37 and Flag-GPR37L1 was assessed following treatment with 100 nM prosaposin for 30 min (n = 3; all points done in triplicate, *P < 0.05). (C) Primary cortical astrocytes were treated with either no siRNA, a scrambled siRNA, or siRNAs directed against GPR37 and/or GPR37L1. These astrocytes were then stimulated with prosaposin (100 nM, 10 min) and activation of ERK was assessed (n = 3; all points done in duplicate, ***P < 0.001). (D) Quantification of the experiments illustrated in C. (E) Primary cortical astrocytes were exposed to H2O2 and cell death was assessed via measuring the release of lactate dehydrogenase into the media. Prosaposin pretreatment (100 nM) protected the astrocytes from the oxidative stress, but the effects of prosaposin were attenuated when GPR37 and/or GPR37L1 was knocked down via siRNA (n = 3; all points done in triplicate, **P < 0.01).

We next explored the effects of prosaposin on ERK phosphorylation and cell survival in cortical astrocytes. Prosaposin treatment (100 nM) robustly stimulated ERK phosphorylation in primary astrocyte cultures, and this response was not affected by pretreatment of the astrocytes with either scrambled siRNA or siRNA directed against GPR37L1 (Fig. 4 C and D). However, siRNA knockdown of GPR37, as well as joint knockdown of both GPR37 and GPR37L1, completely abolished prosaposin-induced ERK phosphorylation. Finally, pretreatment with prosaposin protected cortical astrocytes from H2O2-induced cell death, with knockdown of GPR37, GPR37L1, or GPR37 and GPR37L1 together substantially attenuating the protective effects of the full-length prosaposin (Fig. 4E), similar to findings made earlier regarding the effects of prosaptide on astrocyte survival.

Discussion

The studies presented here have identified prosaposin and prosaptide, a peptide derived from prosaposin, as ligands for the orphan receptors GPR37 and GPR37L1. These ligands bind to GPR37 and GPR37L1, induce receptor internalization, and stimulate GPR37- and GPR37L1-mediated signaling through PTX-sensitive G proteins. For nearly two decades, it has been known that prosaposin and prosaptide can activate G protein pathways, but the putative receptor(s) mediating these effects have eluded identification. We propose that GPR37 and GPR37L1 mediate at least some and potentially all of these G protein-dependent actions of prosaposin and prosaptide, given the striking similarities between the previously defined properties of the putative prosaposin receptors and the findings reported here for GPR37 and GPR37L1. These properties include binding to prosaposin and prosaptide with nanomolar affinity (21, 25, 26, 53), coupling to PTX-sensitive G proteins (25, 28, 29, 35, 37, 43, 55), stimulation of ERK phosphorylation as a downstream readout (26, 28, 5355), and protection against cellular stress (27, 30, 31, 38, 41, 42, 55). As demonstrated by our data, all of these criteria are met by GPR37 and GPR37L1. Moreover, the distribution patterns of GPR37 and GPR37L1 correspond exceptionally well to the expected distribution patterns of the putative prosaposin receptors. In addition to the effects of prosaposin and prosaptide on astrocytes demonstrated in this paper, prosaptide and prosaposin have been shown to exert effects on Schwann cells (26, 28, 59), oligodendrocytes (26), and certain populations of neurons (30, 38, 41, 60), cell types that are known to express significant levels of GPR37 and GPR37L1 (10, 56). Furthermore, it is interesting to note that prosaptide and the invertebrate peptide HA, which has previously been reported to activate GPR37 (17, 18), are similar in length and exhibit significant sequence similarity in their C-terminal regions (K-V-I-L for HA vs. K-E-I-L for prosaptide). HA does not seem to be a true ortholog of prosaptide, but nonetheless we propose that this invertebrate peptide may possess the ability to act as a GPR37 agonist owing to its similarity to prosaptide and prosaposin.

GPR37 and GPR37L1 were found in our studies to be both necessary and sufficient for mediating G protein-dependent signaling by prosaptide and prosaposin in the cell types that were examined. HEK-293T cells, for example, do not show any endogenous responses to prosaptide or prosaposin, and these ligands were found to only be capable of stimulating G protein-mediated signaling in HEK-293T cells when the cells were transfected with either GPR37 or GPR37L1. These experiments reveal that either GPR37 or GPR37L1 is sufficient to confer G protein-mediated signaling by prosaptide and prosaposin to HEK-293T cells. Furthermore, primary astrocytes exhibit robust endogenous increases in phospho-ERK when treated with prosaptide or prosaposin, and these effects are dramatically attenuated by siRNA-mediated knockdown of GPR37. These experiments reveal that, at least in primary astrocytes, GPR37 is necessary for stimulation of ERK by prosaptide and prosaposin. With regard to the abilities of prosaptide and prosaposin to protect primary astrocytes from H2O2-induced cell death, it is interesting to note that knockdown of either GPR37 or GPR37L1 attenuated the protective actions of prosaptide and prosaposin. This difference in findings between the ERK phosphorylation experiments (where only GPR37 was necessary) and the oxidative stress protection studies (where both GPR37 and GPR37L1 were necessary) suggests that GPR37 and GPR37L1 do not couple to exactly the same set of intracellular signaling pathways and that the protective actions of prosaptide and prosaposin via GPR37/GPR37L1 are dependent on more than just downstream activation of ERK. Perhaps future studies will illuminate the receptor-specific differences between GPR37 and GPR37L1, providing insights into why there seem to be at least two distinct receptors for prosaposin in the CNS.

Prosaposin is an interesting multifunctional protein (61). It can be targeted to lysosomes and cleaved into saposins A–D, which play specific roles in the degradation of sphingolipids (61, 62). Alternatively, prosaposin can be released from cells as an intact protein, which acts as a neuroprotective and glioprotective factor (21, 41, 42, 63). The region of prosaposin required for activating G protein-mediated signaling has been narrowed down to a 14-amino acid sequence in the saposin C region of prosaposin, and this peptide fragment has come to be known as prosaptide (23). Interestingly, knockout mice lacking either full-length prosaposin or the saposin C region exhibit striking patterns of neurodegeneration and loss of motor function, although it is not yet clear which aspects of these phenotypes are due to loss of the lysosomal function of the cleaved saposins and which are due to loss of the neuroprotective and glioprotective actions of secreted full-length prosaposin (6468). Loss of prosaposin in humans also results in a dramatic phenotype, including profound neurodegeneration in the cerebral cortex and other brain regions (69, 70), indicating a prominent role for prosaposin in promoting neuronal survival in humans.

Our proposal that GPR37 and GPR37L1 represent receptors for the neuroprotective and glioprotective actions of prosaposin might seem upon first consideration to be at odds with much of the literature on GPR37, which has focused on the propensity of overexpressed GPR37 to induce neurodegeneration (9, 12, 7174). However, it is important to appreciate that published studies linking GPR37 to neurodegeneration have been performed in cells where parkin was knocked out (71, 72) or where GPR37 was highly overexpressed to artificially overwhelm cellular trafficking machinery (9, 12, 73, 74). Parkin is an E3 ubiquitin ligase necessary for targeting misfolded substrates for degradation (11). If parkin is inactivated and its misfolded substrates are allowed to build up and aggregate, this can induce endoplasmic reticulum stress and ultimately cell death (11). Thus, although overexpression of GPR37 can be deleterious in certain cell types if parkin function is lost owing to mutation or knockdown, we propose that GPR37 and GPR37L1 play predominantly protective roles in neurons and glia under normal conditions where parkin is expressed at physiological levels, because our findings provide evidence that GPR37 and GPR37L1 mediate protective actions of secreted prosaposin. Consistent with this idea, GPR37 was recently shown to exert striking protective actions in neuron-like N2a cells, an observation that fits well with our proposal that GPR37 exerts mainly protective effects in cells where the receptor is properly folded and trafficked (75).

Future work may determine the precise roles that GPR37 and/or GPR37L1 play in mediating the various in vivo physiological effects that have been reported for prosaposin and prosaptide. In pursuing such in vivo studies, it is useful to consider a model of the potential physiological significance of this signaling system. It has been demonstrated that the expression of prosaposin is strikingly up-regulated following nerve injury (76). Conceivably, activation of GPR37 and/or GPR37L1 by up-regulated prosaposin following injury could prompt neurons and glia to initiate intracellular signaling pathways that promote survival in the face of the cellular stress associated with injury and inflammation. If future studies support this model, then GPR37 and GPR37L1 may eventually serve as outstanding targets for novel therapeutics aimed at enhancing neuronal and glial survival in response to injury. Prosaposin and prosaptide have been shown to protect dopaminergic neurons in animal models of Parkinson disease (38, 41), facilitate neuronal survival following focal cerebral ischemia (22, 32), reduce neuropathic pain (37, 40, 57), and promote remyelination of nerves following injury (26, 28, 59). Thus, if future work in vivo upholds the idea that the neuroprotective and glioprotective actions of prosaposin and prosaptide are mediated by GPR37 and GPR37L1, the development of small molecule ligands for these receptors may provide new therapeutic possibilities for the treatment of Parkinson disease, stroke, neuropathic pain, and myelination disorders.

Materials and Methods

Detailed materials and methods are given in SI Materials and Methods.

Cell Culture.

Cells were cultured as described previously (44). Cortical astrocytes were derived from mice 1–2 d old and grown for 7–10 d in culture.

Plasmids, Antibodies, and Peptides.

SI Materials and Methods gives the origins of antibodies, constructs, and peptides.

siRNA Knockdown.

The siRNAs directed against GPR37 and GPR37L1 were incorporated into primary cortical astrocytes. Cells were harvested 72 h following nucleofection.

Prosaposin Production.

Prosaposin was expressed and purified using a slightly modified version of a previously published protocol (58).

Western Blotting.

Samples were processed and run on gels as previously described (44).

ERK Phosphorylation Assay.

Cells were serum-starved for 2–3 h before assay stimulation. Cells were incubated with ligand for 10 min in DMEM at 37 °C.

Cell Surface Luminometry.

Assays of receptor surface expression were performed as previously described (44).

cAMP Assay.

Transfected HEK-293T cells treated with forskolin and/or prosaptide. Samples were analyzed using a colorimetric cAMP ELISA kit (Cell Biolabs).

Biotin Binding and Pulldown.

Biotinylated prosaptide TX14(A) was attached to Neutravadin beads. Soluble lysates were incubated with beads and the resulting samples were examined via Western blot for pulldown of solubilized receptors.

35S-GTPγS Binding.

Membranes derived from transfected HEK-293T cells were incubated with 35S-GTPγS and binding was assessed using a scintillation counter.

Microscopy.

Transfected COS-7 cells were incubated with rabbit anti-FLAG antibodies, then treated and fixed. Surface-expressed receptors were labeled red and internalized receptors were labeled green.

Cytotoxicity.

Forty-eight hours following nucleofection, cells were treated with 500 µM H2O2 in DMEM with or without drug for 24 h. Media was harvested and analyzed via a cytotoxicity kit (Promega).

Supplementary Material

Supporting Information

Acknowledgments

We thank Stefanie Ritter (Emory University) for technical advice and Thomas Kukar and his laboratory (Emory University) for the use of imaging equipment. This work was supported by National Institutes of Health Grant R21-NS081461 and Pilot Project Award P01-ES016731 from the Emory Parkinson’s Disease Collaborative Environmental Research Center. R.C.M. was supported by National Institute on Drug Abuse Training Grant T32-DA015040.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1219004110/-/DCSupplemental.

References

  • 1.Lagerström MC, Schiöth HB. Structural diversity of G protein-coupled receptors and significance for drug discovery. Nat Rev Drug Discov. 2008;7(4):339–357. doi: 10.1038/nrd2518. [DOI] [PubMed] [Google Scholar]
  • 2.Civelli O, Saito Y, Wang Z, Nothacker HP, Reinscheid RK. Orphan GPCRs and their ligands. Pharmacol Ther. 2006;110(3):525–532. doi: 10.1016/j.pharmthera.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 3.Marazziti D, et al. Cloning of GPR37, a gene located on chromosome 7 encoding a putative G-protein-coupled peptide receptor, from a human frontal brain EST library. Genomics. 1997;45(1):68–77. doi: 10.1006/geno.1997.4900. [DOI] [PubMed] [Google Scholar]
  • 4.Zeng Z, Su K, Kyaw H, Li Y. A novel endothelin receptor type-B-like gene enriched in the brain. Biochem Biophys Res Commun. 1997;233(2):559–567. doi: 10.1006/bbrc.1997.6408. [DOI] [PubMed] [Google Scholar]
  • 5.Donohue PJ, et al. A human gene encodes a putative G protein-coupled receptor highly expressed in the central nervous system. Brain Res Mol Brain Res. 1998;54(1):152–160. doi: 10.1016/s0169-328x(97)00336-7. [DOI] [PubMed] [Google Scholar]
  • 6.Marazziti D, Gallo A, Golini E, Matteoni R, Tocchini-Valentini GP. Molecular cloning and chromosomal localization of the mouse Gpr37 gene encoding an orphan G-protein-coupled peptide receptor expressed in brain and testis. Genomics. 1998;53(3):315–324. doi: 10.1006/geno.1998.5433. [DOI] [PubMed] [Google Scholar]
  • 7.Valdenaire O, et al. A new family of orphan G protein-coupled receptors predominantly expressed in the brain. FEBS Lett. 1998;424(3):193–196. doi: 10.1016/s0014-5793(98)00170-7. [DOI] [PubMed] [Google Scholar]
  • 8.Leng N, Gu G, Simerly RB, Spindel ER. Molecular cloning and characterization of two putative G protein-coupled receptors which are highly expressed in the central nervous system. Brain Res Mol Brain Res. 1999;69(1):73–83. doi: 10.1016/s0169-328x(99)00092-3. [DOI] [PubMed] [Google Scholar]
  • 9.Imai Y, et al. An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress, is a substrate of Parkin. Cell. 2001;105(7):891–902. doi: 10.1016/s0092-8674(01)00407-x. [DOI] [PubMed] [Google Scholar]
  • 10.Cahoy JD, et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci. 2008;28(1):264–278. doi: 10.1523/JNEUROSCI.4178-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Walden H, Martinez-Torres RJ. Regulation of Parkin E3 ubiquitin ligase activity. Cell Mol Life Sci. 2012;69(18):3053–3067. doi: 10.1007/s00018-012-0978-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yang Y, Nishimura I, Imai Y, Takahashi R, Lu B. Parkin suppresses dopaminergic neuron-selective neurotoxicity induced by Pael-R in Drosophila. Neuron. 2003;37(6):911–924. doi: 10.1016/s0896-6273(03)00143-0. [DOI] [PubMed] [Google Scholar]
  • 13.Obeso JA, et al. Missing pieces in the Parkinson’s disease puzzle. Nat Med. 2010;16(6):653–661. doi: 10.1038/nm.2165. [DOI] [PubMed] [Google Scholar]
  • 14.Marazziti D, et al. Altered dopamine signaling and MPTP resistance in mice lacking the Parkinson’s disease-associated GPR37/parkin-associated endothelin-like receptor. Proc Natl Acad Sci USA. 2004;101(27):10189–10194. doi: 10.1073/pnas.0403661101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Imai Y, et al. Pael receptor is involved in dopamine metabolism in the nigrostriatal system. Neurosci Res. 2007;59(4):413–425. doi: 10.1016/j.neures.2007.08.005. [DOI] [PubMed] [Google Scholar]
  • 16.Marazziti D, et al. GPR37 associates with the dopamine transporter to modulate dopamine uptake and behavioral responses to dopaminergic drugs. Proc Natl Acad Sci USA. 2007;104(23):9846–9851. doi: 10.1073/pnas.0703368104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rezgaoui M, et al. The neuropeptide head activator is a high-affinity ligand for the orphan G-protein-coupled receptor GPR37. J Cell Sci. 2006;119(Pt 3):542–549. doi: 10.1242/jcs.02766. [DOI] [PubMed] [Google Scholar]
  • 18.Gandía J, et al. The Parkinson’s disease-associated GPR37 receptor-mediated cytotoxicity is controlled by its intracellular cysteine-rich domain. J Neurochem. 2013;125(3):362–372. doi: 10.1111/jnc.12196. [DOI] [PubMed] [Google Scholar]
  • 19.Bodenmüller H, Schaller HC, Darai G. Human hypothalamus and intestine contain a hydra-neuropeptide. Neurosci Lett. 1980;16(1):71–74. doi: 10.1016/0304-3940(80)90103-2. [DOI] [PubMed] [Google Scholar]
  • 20.Bodenmüller H, Schaller HC. Conserved amino acid sequence of a neuropeptide, the head activator, from coelenterates to humans. Nature. 1981;293(5833):579–580. doi: 10.1038/293579a0. [DOI] [PubMed] [Google Scholar]
  • 21.O’Brien JS, Carson GS, Seo HC, Hiraiwa M, Kishimoto Y. Identification of prosaposin as a neurotrophic factor. Proc Natl Acad Sci USA. 1994;91(20):9593–9596. doi: 10.1073/pnas.91.20.9593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sano A, et al. Protection by prosaposin against ischemia-induced learning disability and neuronal loss. Biochem Biophys Res Commun. 1994;204(2):994–1000. doi: 10.1006/bbrc.1994.2558. [DOI] [PubMed] [Google Scholar]
  • 23.O’Brien JS, et al. Identification of the neurotrophic factor sequence of prosaposin. FASEB J. 1995;9(8):681–685. doi: 10.1096/fasebj.9.8.7768361. [DOI] [PubMed] [Google Scholar]
  • 24.Kotani Y, et al. A hydrophilic peptide comprising 18 amino acid residues of the prosaposin sequence has neurotrophic activity in vitro and in vivo. J Neurochem. 1996;66(5):2197–2200. doi: 10.1046/j.1471-4159.1996.66052197.x. [DOI] [PubMed] [Google Scholar]
  • 25.Hiraiwa M, Campana WM, Martin BM, O’Brien JS. Prosaposin receptor: Evidence for a G-protein-associated receptor. Biochem Biophys Res Commun. 1997;240(2):415–418. doi: 10.1006/bbrc.1997.7673. [DOI] [PubMed] [Google Scholar]
  • 26.Hiraiwa M, Taylor EM, Campana WM, Darin SJ, O’Brien JS. Cell death prevention, mitogen-activated protein kinase stimulation, and increased sulfatide concentrations in Schwann cells and oligodendrocytes by prosaposin and prosaptides. Proc Natl Acad Sci USA. 1997;94(9):4778–4781. doi: 10.1073/pnas.94.9.4778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Campana WM, et al. Prosaptide prevents paclitaxel neurotoxicity. Neurotoxicology. 1998;19(2):237–244. [PubMed] [Google Scholar]
  • 28.Campana WM, Hiraiwa M, O’Brien JS. Prosaptide activates the MAPK pathway by a G-protein-dependent mechanism essential for enhanced sulfatide synthesis by Schwann cells. FASEB J. 1998;12(3):307–314. doi: 10.1096/fasebj.12.3.307. [DOI] [PubMed] [Google Scholar]
  • 29.Misasi R, et al. Colocalization and complex formation between prosaposin and monosialoganglioside GM3 in neural cells. J Neurochem. 1998;71(6):2313–2321. doi: 10.1046/j.1471-4159.1998.71062313.x. [DOI] [PubMed] [Google Scholar]
  • 30.Tsuboi K, Hiraiwa M, O’Brien JS. Prosaposin prevents programmed cell death of rat cerebellar granule neurons in culture. Brain Res Dev Brain Res. 1998;110(2):249–255. doi: 10.1016/s0165-3806(98)00109-6. [DOI] [PubMed] [Google Scholar]
  • 31.Campana WM, Darin SJ, O’Brien JS. Phosphatidylinositol 3-kinase and Akt protein kinase mediate IGF-I- and prosaptide-induced survival in Schwann cells. J Neurosci Res. 1999;57(3):332–341. [PubMed] [Google Scholar]
  • 32.Igase K, et al. An 18-mer peptide fragment of prosaposin ameliorates place navigation disability, cortical infarction, and retrograde thalamic degeneration in rats with focal cerebral ischemia. J Cereb Blood Flow Metab. 1999;19(3):298–306. doi: 10.1097/00004647-199903000-00008. [DOI] [PubMed] [Google Scholar]
  • 33.Campana WM, Mohiuddin L, Misasi R, O’Brien JS, Calcutt NA. Prosaposin-derived peptides enhanced sprouting of sensory neurons in vitro and induced sprouting at motor endplates in vivo. J Peripher Nerv Syst. 2000;5(3):126–130. doi: 10.1046/j.1529-8027.2000.00019.x. [DOI] [PubMed] [Google Scholar]
  • 34.Lu AG, Otero DA, Hiraiwa M, O’Brien JS. Neuroprotective effect of retro-inverso Prosaptide D5 on focal cerebral ischemia in rat. Neuroreport. 2000;11(8):1791–1794. doi: 10.1097/00001756-200006050-00039. [DOI] [PubMed] [Google Scholar]
  • 35.Taylor EM, Otero DA, Banks WA, O’Brien JS. Retro-inverso prosaptide peptides retain bioactivity, are stable In vivo, and are blood-brain barrier permeable. J Pharmacol Exp Ther. 2000;295(1):190–194. [PubMed] [Google Scholar]
  • 36.Taylor EM, Otero DA, Banks WA, O’Brien JS. Designing stable blood-brain barrier-permeable prosaptide peptides for treatment of central nervous system neurodegeneration. J Pharmacol Exp Ther. 2000;293(2):403–409. [PubMed] [Google Scholar]
  • 37.Yan L, Otero DA, Hiraiwa M, O’Brien JS. Prosaptide D5 reverses hyperalgesia: inhibition of calcium channels through a pertussis toxin-sensitive G-protein mechanism in the rat. Neurosci Lett. 2000;278(1–2):120–122. doi: 10.1016/s0304-3940(99)00902-7. [DOI] [PubMed] [Google Scholar]
  • 38.Liu J, Wang CY, O’Brien JS. Prosaptide D5, a retro-inverso 11-mer peptidomimetic, rescued dopaminergic neurons in a model of Parkinson’s disease. FASEB J. 2001;15(6):1080–1082. doi: 10.1096/fj.00-0603fje. [DOI] [PubMed] [Google Scholar]
  • 39.Morita F, et al. Protective effect of a prosaposin-derived, 18-mer peptide on slowly progressive neuronal degeneration after brief ischemia. J Cereb Blood Flow Metab. 2001;21(11):1295–1302. doi: 10.1097/00004647-200111000-00005. [DOI] [PubMed] [Google Scholar]
  • 40.Jolivalt CG, Vu Y, Mizisin LM, Mizisin AP, Calcutt NA. Impaired prosaposin secretion during nerve regeneration in diabetic rats and protection of nerve regeneration by a prosaposin-derived peptide. J Neuropathol Exp Neurol. 2008;67(7):702–710. doi: 10.1097/NEN.0b013e31817e23f4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ochiai T, et al. Molecular mechanism for neuro-protective effect of prosaposin against oxidative stress: Its regulation of dimeric transcription factor formation. Biochim Biophys Acta. 2008;1780(12):1441–1447. doi: 10.1016/j.bbagen.2008.07.010. [DOI] [PubMed] [Google Scholar]
  • 42.Li N, Sarojini H, An J, Wang E. Prosaposin in the secretome of marrow stroma-derived neural progenitor cells protects neural cells from apoptotic death. J Neurochem. 2010;112(6):1527–1538. doi: 10.1111/j.1471-4159.2009.06565.x. [DOI] [PubMed] [Google Scholar]
  • 43.Lee TJ, Sartor O, Luftig RB, Koochekpour S. Saposin C promotes survival and prevents apoptosis via PI3K/Akt-dependent pathway in prostate cancer cells. Mol Cancer. 2004;3:31. doi: 10.1186/1476-4598-3-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dunham JH, Meyer RC, Garcia EL, Hall RA. GPR37 surface expression enhancement via N-terminal truncation or protein-protein interactions. Biochemistry. 2009;48(43):10286–10297. doi: 10.1021/bi9013775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.von Zastrow M. Role of endocytosis in signalling and regulation of G-protein-coupled receptors. Biochem Soc Trans. 2001;29(Pt 4):500–504. doi: 10.1042/bst0290500. [DOI] [PubMed] [Google Scholar]
  • 46.Gasser MC, Berti I, Hauser KF, Fischer-Colbrie R, Saria A. Secretoneurin promotes pertussis toxin-sensitive neurite outgrowth in cerebellar granule cells. J Neurochem. 2003;85(3):662–669. doi: 10.1046/j.1471-4159.2003.01677.x. [DOI] [PubMed] [Google Scholar]
  • 47.Igwe OJ, Kim DC, Seybold VS, Larson AA. Specific binding of substance P aminoterminal heptapeptide [SP(1-7)] to mouse brain and spinal cord membranes. J Neurosci. 1990;10(11):3653–3663. doi: 10.1523/JNEUROSCI.10-11-03653.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lakatos A, Prinster S, Vicentic A, Hall RA, Kuhar MJ. Cocaine- and amphetamine-regulated transcript (CART) peptide activates the extracellular signal-regulated kinase (ERK) pathway in AtT20 cells via putative G-protein coupled receptors. Neurosci Lett. 2005;384(1-2):198–202. doi: 10.1016/j.neulet.2005.04.072. [DOI] [PubMed] [Google Scholar]
  • 49.Lin Y, Hall RA, Kuhar MJ. CART peptide stimulation of G protein-mediated signaling in differentiated PC12 cells: Identification of PACAP 6-38 as a CART receptor antagonist. Neuropeptides. 2011;45(5):351–358. doi: 10.1016/j.npep.2011.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chiu S, Wong YW, Wan YP, Chiu P, Mishra RK. Are the pharmacological effects of L-prolyl-L-leucyl-glycinamide (PLG) mediated through specific receptor mechanisms? Prog Neuropsychopharmacol Biol Psychiatry. 1983;7(4-6):739–742. doi: 10.1016/0278-5846(83)90056-8. [DOI] [PubMed] [Google Scholar]
  • 51.Akiyama N, et al. Biotin derivatives of endothelin: Utilization for affinity purification of endothelin receptor. Protein Expr Purif. 1992;3(5):427–433. doi: 10.1016/s1046-5928(05)80046-4. [DOI] [PubMed] [Google Scholar]
  • 52.Saravanan K, Paramasivam M, Dey S, Singh TP, Srinivasan A. Biotinyl endothelin-1 binding to endothelin receptor and its applications. J Cardiovasc Pharmacol. 2004;44(3):287–293. doi: 10.1097/01.fjc.0000132316.24513.ec. [DOI] [PubMed] [Google Scholar]
  • 53.Campana WM, Hiraiwa M, Addison KC, O’Brien JS. Induction of MAPK phosphorylation by prosaposin and prosaptide in PC12 cells. Biochem Biophys Res Commun. 1996;229(3):706–712. doi: 10.1006/bbrc.1996.1869. [DOI] [PubMed] [Google Scholar]
  • 54.Hiraiwa M, Campana WM, Wang CY, Otero DA, O’Brien JS. A retro-inverso Prosaptide D5 promotes a myelination process in developing rats. Brain Res Dev Brain Res. 2001;128(1):73–76. doi: 10.1016/s0165-3806(01)00141-9. [DOI] [PubMed] [Google Scholar]
  • 55.Misasi R, et al. Prosaposin treatment induces PC12 entry in the S phase of the cell cycle and prevents apoptosis: Activation of ERKs and sphingosine kinase. FASEB J. 2001;15(2):467–474. doi: 10.1096/fj.00-0217com. [DOI] [PubMed] [Google Scholar]
  • 56.Galey D, et al. Differential transcriptional regulation by human immunodeficiency virus type 1 and gp120 in human astrocytes. J Neurovirol. 2003;9(3):358–371. doi: 10.1080/13550280390201119. [DOI] [PubMed] [Google Scholar]
  • 57.Calcutt NA, et al. Prosaposin gene expression and the efficacy of a prosaposin-derived peptide in preventing structural and functional disorders of peripheral nerve in diabetic rats. J Neuropathol Exp Neurol. 1999;58(6):628–636. doi: 10.1097/00005072-199906000-00007. [DOI] [PubMed] [Google Scholar]
  • 58.Xu C, et al. The interaction of the second Kunitz-type domain (KD2) of TFPI-2 with a novel interaction partner, prosaposin, mediates the inhibition of the invasion and migration of human fibrosarcoma cells. Biochem J. 2012;441(2):665–674. doi: 10.1042/BJ20110533. [DOI] [PubMed] [Google Scholar]
  • 59.Hiraiwa M, Campana WM, Mizisin AP, Mohiuddin L, O’Brien JS. Prosaposin: A myelinotrophic protein that promotes expression of myelin constituents and is secreted after nerve injury. Glia. 1999;26(4):353–360. [PubMed] [Google Scholar]
  • 60.Cove J, Morales CR, Baranes D. SGP-1 increases dendritic and synaptic development dependent on synaptic activity. Neurosci Res. 2006;56(4):372–385. doi: 10.1016/j.neures.2006.08.008. [DOI] [PubMed] [Google Scholar]
  • 61.Misasi R, et al. Biochemistry and neurobiology of prosaposin: A potential therapeutic neuro-effector. Cent Nerv Syst Agents Med Chem. 2009;9(2):119–131. doi: 10.2174/187152409788452045. [DOI] [PubMed] [Google Scholar]
  • 62.Kishimoto Y, Hiraiwa M, O’Brien JS. Saposins: Structure, function, distribution, and molecular genetics. J Lipid Res. 1992;33(9):1255–1267. [PubMed] [Google Scholar]
  • 63.Hineno T, et al. Secretion of sphingolipid hydrolase activator precursor, prosaposin. Biochem Biophys Res Commun. 1991;176(2):668–674. doi: 10.1016/s0006-291x(05)80236-0. [DOI] [PubMed] [Google Scholar]
  • 64.Fujita N, et al. Targeted disruption of the mouse sphingolipid activator protein gene: A complex phenotype, including severe leukodystrophy and wide-spread storage of multiple sphingolipids. Hum Mol Genet. 1996;5(6):711–725. doi: 10.1093/hmg/5.6.711. [DOI] [PubMed] [Google Scholar]
  • 65.Oya Y, Nakayasu H, Fujita N, Suzuki K, Suzuki K. Pathological study of mice with total deficiency of sphingolipid activator proteins (SAP knockout mice) Acta Neuropathol. 1998;96(1):29–40. doi: 10.1007/s004010050857. [DOI] [PubMed] [Google Scholar]
  • 66.Sun Y, et al. Combined saposin C and D deficiencies in mice lead to a neuronopathic phenotype, glucosylceramide and alpha-hydroxy ceramide accumulation, and altered prosaposin trafficking. Hum Mol Genet. 2007;16(8):957–971. doi: 10.1093/hmg/ddm040. [DOI] [PubMed] [Google Scholar]
  • 67.Sun Y, et al. Specific saposin C deficiency: CNS impairment and acid beta-glucosidase effects in the mouse. Hum Mol Genet. 2010;19(4):634–647. doi: 10.1093/hmg/ddp531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yoneshige A, Suzuki K, Suzuki K, Matsuda J. A mutation in the saposin C domain of the sphingolipid activator protein (Prosaposin) gene causes neurodegenerative disease in mice. J Neurosci Res. 2010;88(10):2118–2134. doi: 10.1002/jnr.22371. [DOI] [PubMed] [Google Scholar]
  • 69.Elleder M, et al. Prosaposin deficiency—A rarely diagnosed, rapidly progressing, neonatal neurovisceral lipid storage disease. Report of a further patient. Neuropediatrics. 2005;36(3):171–180. doi: 10.1055/s-2005-865608. [DOI] [PubMed] [Google Scholar]
  • 70.Sikora J, Harzer K, Elleder M. Neurolysosomal pathology in human prosaposin deficiency suggests essential neurotrophic function of prosaposin. Acta Neuropathol. 2007;113(2):163–175. doi: 10.1007/s00401-006-0148-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kitao Y, et al. Pael receptor induces death of dopaminergic neurons in the substantia nigra via endoplasmic reticulum stress and dopamine toxicity, which is enhanced under condition of parkin inactivation. Hum Mol Genet. 2007;16(1):50–60. doi: 10.1093/hmg/ddl439. [DOI] [PubMed] [Google Scholar]
  • 72.Wang HQ, et al. Pael-R transgenic mice crossed with parkin deficient mice displayed progressive and selective catecholaminergic neuronal loss. J Neurochem. 2008;107(1):171–185. doi: 10.1111/j.1471-4159.2008.05607.x. [DOI] [PubMed] [Google Scholar]
  • 73.Dusonchet J, Bensadoun JC, Schneider BL, Aebischer P. Targeted overexpression of the parkin substrate Pael-R in the nigrostriatal system of adult rats to model Parkinson’s disease. Neurobiol Dis. 2009;35(1):32–41. doi: 10.1016/j.nbd.2009.03.013. [DOI] [PubMed] [Google Scholar]
  • 74.Marazziti D, et al. Induction of macroautophagy by overexpression of the Parkinson’s disease-associated GPR37 receptor. FASEB J. 2009;23(6):1978–1987. doi: 10.1096/fj.08-121210. [DOI] [PubMed] [Google Scholar]
  • 75.Lundius EG, Stroth N, Vukojević V, Terenius L, Svenningsson P. Functional GPR37 trafficking protects against toxicity induced by 6-OHDA, MPP+ or rotenone in a catecholaminergic cell line. J Neurochem. 2013;124(3):410–417. doi: 10.1111/jnc.12081. [DOI] [PubMed] [Google Scholar]
  • 76.Chen J, et al. Expression patterns in alternative splicing forms of prosaposin mRNA in the rat facial nerve nucleus after facial nerve transection. Neurosci Res. 2008;60(1):82–94. doi: 10.1016/j.neures.2007.09.010. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES