Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 1.
Published in final edited form as: Trends Pharmacol Sci. 2019 Aug 3;40(9):636–654. doi: 10.1016/j.tips.2019.07.006

The molecular function of σ receptors: past, present, and future

Hayden R Schmidt 1,2,*, Andrew C Kruse 1,*
PMCID: PMC6748033  NIHMSID: NIHMS1534843  PMID: 31387763

Abstract

The σ1 and σ2 receptors are enigmatic proteins that have attracted attention for decades due to the chemical diversity and therapeutic potential of their ligands. However, despite ongoing clinical trials with σ receptor ligands for multiple conditions, relatively little is known regarding the molecular function of these receptors. In this review, we revisit past research on σ receptors, and discuss the interpretation of these data in light of recent developments. We provide a synthesis of emerging structural and genetic data on the σ1 receptor and discuss the recent cloning of the σ2 receptor. Finally, we discuss the major questions that remain in the study of σ receptors.

Keywords: σ1 receptor, σ2 receptor/TMEM97, structural pharmacology, molecular pharmacology

The σ receptors: enigmatic therapeutic targets

The σ1 and σ2 receptors have been the subject of intense study by pharmacologists for over four decades [1, 2]. Both receptors have been proposed as therapeutic targets for several diseases and conditions. The σ1 receptor is considered a potential therapeutic target for pain management [3] and neurological pathologies such as amyotrophic lateral sclerosis (ALS) [4, 5], Alzheimer’s disease [4, 5], Parkinson’s disease [4, 5], retinal disease [6], stroke [4], and cocaine [7] and alcohol [8] addiction. Additionally, there is interest in using σ1 and σ2 receptor ligands for treating [9, 10] and imaging [11] cancer. Currently, σ1 receptor ligands are in clinical trials for the treatment of chemotherapy-induced neuropathic pain [12], Alzheimer’s disease [13], and ischemic stroke [14]. Meanwhile, one σ2 receptor ligand recently showed efficacy against the negative symptoms of schizophrenia in a phase II clinical trial [15], and another was well tolerated in a phase I clinical trial for Alzheimer’s disease and is now entering phase II [16].

Despite intense therapeutic interest, many of the molecular details of both σ1 and σ2 receptor functions remain unclear. The last five years have seen considerable progress in σ receptor genetics, structural biology, and biochemistry, but major questions remain unanswered. In this review we discuss recent advances in σ receptor molecular biology and biochemistry and consider both how these advances inform our interpretation of previous work and what major challenges lie ahead.

Discovery and molecular pharmacology of the σ receptors

In 1976, pharmacological studies of opioids and opioid-like compounds in dogs led to the proposal of three distinct opioid receptor subtypes: μ, κ, and σ [1]. However, radioligand binding experiments quickly revealed that the σ receptor binding site was distinct from the opioid receptors [1]. Specifically, the σ receptor does not bind to classical opioid ligands such as naloxone, etorphine, or (−) benzomorphans [1]. Instead, the σ receptor has high affinity for (+) benzomorphans [1] in addition to myriad small molecules that exhibit little structural similarity to one another [1] (Figure 1). The receptor’s unusual pharmacological profile attracted interest from pharmacologists throughout the 1980s and 1990s. However, the receptor’s promiscuous ligand binding profile meant that few selective ligands existed, complicating efforts to unambiguously ascribe pharmacological effects to it. This difficulty was overcome with the development of [3H](+)-pentazocine, a radioligand with high affinity and specificity for the σ1 receptor [17].

Figure 1: Representative σ receptor ligands and the central pharmacophore.

Figure 1:

A depiction of some high-affinity σ receptor ligands, as well as the central σ1 receptor pharmacophore. Adapted from Glennon et al., 2005 [19].

The use of [3H](+)-pentazocine enabled two major advances in σ receptor pharmacology. The first was the identification of two distinct σ receptor binding sites [18]. The first site was dubbed σ1 and largely corresponds to the classical σ receptor defined by Su and Tam [1] described above. The second site was named σ2, and like σ1 it exhibits high affinity for both ditolylguanidine (DTG) and haloperidol. However, the σ2 receptor does not bind benzomorphans [18]. The second major advance enabled by [3H](+)-pentazocine was the identification of a minimal pharmacophore (see Glossary) sufficient for high-affinity binding to the σ1 receptor. This simple pharmacophore features a single positively charged nitrogen flanked by two hydrophobic or aromatic moieties 6 – 10 Å and 2.5 – 3.9 Å in length [19] (Figure 1). All known σ1 receptor ligands with high affinity (KD < 50 nM) fit this model [19].

No endogenous ligand has been definitively identified for σ receptors. Early work demonstrated that the σ1 receptor has affinity for some steroids, especially progesterone [1]. However, physiological concentrations of progesterone are thought to be low relative to its Kd for the σ1 receptor [20], and though the σ1 receptor is sometimes localized to the plasma membrane, it is primarily an intracellular receptor [21]. However, pharmacological manipulation of steroid synthesis can alter the accessibility of σ1 receptor sites in the brain [22]. Similarly, others have proposed that the hallucinogen N,N-dimethyltryptamine (DMT) is an endogenous ligand for the σ receptor [23], but DMT has only a 14.75 μM affinity for the σ1 receptor, while its plasma concentrations are not thought to exceed 500 nM, making it unlikely that this interaction is physiological [24]. Additionally, other work has shown that DMT has much higher affinity for 5-HT receptors, which are probably responsible for its hallucinogenic effects [25]. D-erythro-sphingosine has also been proposed as an endogenous σ1 receptor ligand, though the affinity for the receptor was variable depending on the assay used [26], and it has not been demonstrated that the interaction occurs in living cells. A recent paper suggested that choline may serve as an endogenous ligand for the σ1 receptor [27], raising another possibility.

Molecular function of the σ1 receptor

Despite over 40 years of study, there is still much to be learned about the molecular role of the σ1 receptor in cells. A prevailing model is that the σ1 receptor modulates other cellular signaling pathways by acting as a ligand-operated chaperone. Ligands of the σ1 receptor have historically been classified as agonists or antagonists based on their ability to recapitulate the effects of genetic knockout or knockdown of the σ1 receptor, typically in animal models [4]. Ligands that mimic σ1 receptor genetic knockout are considered antagonists, while ligands that exert some σ1-dependent effect distinct from genetic knockout are considered agonists [4]. A central challenge in functional studies of the σ1 receptor is its lack of similarity to other human proteins. The σ1 receptor was cloned in 1996 [28], and while the σ1 receptor is conserved among vertebrates, it bears no similarity to any other mammalian protein. Its closest homolog is the yeast C8-C7 sterol isomerase, ERG2p [28]. However, the σ1 receptor itself has no sterol isomerase activity [28].

The σ1 receptor as a modulator of cellular signaling

In general, the σ1 receptor is thought of as a modulator of other signaling pathways, particularly G protein-coupled receptor (GPCR) and ion channel signaling. Throughout the 1980s and 1990s, evidence suggested that the σ1 receptor may be involved in intracellular calcium signaling and inositol triphosphate (IP3) turnover [1, 2932]. In 2001, Hayashi and Su used co-immunoprecipitation (Co-IP) experiments to suggest that at least some these effects were mediated through a complex of σ1 receptor, IP3 receptor, and ankyrin B [33] (Table 1). The next year, Aydar et al. showed that σ1 receptor activation could inhibit potassium channels in Xenopus oocytes and that the σ1 receptor could Co-IP with Kv1.4 [34]. These two studies precipitated a shift in the way σ1 receptor was thought to modulate other signaling pathways. Over the last two decades, the σ1 receptor has been shown to influence the cellular function of many proteins, and the proposed mechanism for this modulation has often been direct σ1 receptor-protein interactions (Table 1). To date, the σ1 receptor has been reported to bind to at least 49 proteins, many of which are highly divergent in sequence and structure (Table 1).

Table 1: List of reported experiments positing σ1 receptor protein-protein interactions.

A list of published experiments that have been used to suggest direct protein-protein interactions between the σ1 receptor and other proteins. For constructs, the name of the proteins/tags are listed from N- to C-terminus, such that tags on the N-terminus precede the name of the protein, while tags on the C-terminus proceed the name of the protein. Only interactions reported using low-throughput methods are shown. For the purposes of this table, a “pull down” refers to experiments where at least one of the components was purified. Co-IP: co-immunoprecipitation, BRET: bioluminescence resonance energy transfer, HTRF: homogenous time-resolved fluorescence, SRET: sequential resonance energy transfer, NMR: nuclear magnetic resonance, BiFC: bimolecular fluorescence complementation.

Protein partner Human gene Method σ1 receptor construct Protein partner construct Cell/tissue type Expression method Refs
Ion channels
Inositol triphosphate receptor (IP3R) ITPR3 Co-IP Native σ1 receptor Native IP3R NG-108 cells Native expression [33]
Co-IP Native σ1 receptor Native IP3R CHO cells Native expression [21]
Co-IP σ1 receptor-EGFP Native IP3R NG-108 cells Transient overexpression (σ1 receptor)
Native expression (IP3R)
Co-IP σ1 receptor-EGFP Native IP3R CHO cells Transient overexpression (σ1 receptor)
Native expression (IP3R)
Co-IP Native σ1 receptor Native IP3R Isolated bovine brain mitochondria Native expression [100]
ITPR1 Co-IP Native σ1 receptor Native IP3R NG-108 cells Native expression [101]
Proximity ligation Native σ1 receptor Native IP3R NG-108 cells Native expression
ITPR2 Co-IP Native σ1 receptor Native IP3R Rat heart tissue Native expression [102]
Rynadine receptor 2 (RYR2) RYR2 Co-IP Native σ1 receptor Native RYR2 Rat heart tissue Native expression [102]
Kv1.2 K+ channel KCNA2 Co-IP Native σ1 receptor Native Kv1.2 Mouse nucleus accumbens lysate Native expression [38]
Co-IP with cross-linking σ1 receptor-V5-His Wildtype Kv1.2 NG108–15 cells Transient overexpression
Kv1.3 K+ channel KCNA3 Co-IP σ1 receptor-FLAG Kv1.3-HA HEK 293 cells Transient overexpression [103]
Kv1.4 K+ channel KCNA4 Co-IP Native σ1 receptor Native Kv1.4 Rat posterior pituitary lysate Native expression [34]
L-type voltage-gated calcium channel (VGCC) CACNA1C Co-IP Wildtype σ1 receptor Native L-type VGCC RGC-5 cells Stable overexpression (σ1 receptor in some experiments) [104]
Native expression (VGCC, and σ1 receptor in one experiment)
Acid-sensing ion channel 1a (ASIC1a) ASIC1 Ni affinity chromatography σ1 receptor-FLAG-His ASIC1a-His HEK 293 cells Stable overexpression (ASIC1a)
Transient
[57]
overexpression (σ1 receptor)
Nav1.5 Na+ channel SCN5A Anti-FLAG chromatography σ1 receptor-FLAG Nav1.5-HA tSA 201 cells Transient overexpression [37]
Proximity ligation
Co-IP Native σ1 receptor Native Nav1.5 MDA-MB-468 cells Native expression [35]
Co-IP Native σ1 receptor Native Nav1.5 MDA-MB-231 cells Native expression
N-methyl-D-aspartate receptor (NMDAR) GluN1 subunit GRIN1 Anti-FLAG chromatography σ1 receptor-FLAG Wildtype GluN1 tsA 201 cells Transient overexpression [36]
Anti-FLAG chromatography σ1 receptor-FLAG Wildtype GluN1 NG108–15 cells Transient overexpression
Proximity ligation σ1 receptor-FLAG GluN1-HA tsA 201 cells Transient overexpression
Pull down σ1 receptor-TEV GluN1 c-terminal fragment C0-C1-C2 Purified Protein Bacterial expression and purification [105]
Pull down σ1 receptor-TEV GluN1 c-terminal fragment C0-C1-C2 Purified Protein Bacterial expression and purification [106]
Pull down σ1 receptor-TEV GluN1 c-terminal fragment C0-C1-C2 Purified Protein Bacterial expression and purification [107]
BiFC S1R-split Venus GluN1-split Venus CHO cells Transient overexpression [108]
NMDAR Glun2a subunit GRIN2a Co-IP Native σ1 receptor Native GluN2a Rat hippocampus P2 pellet Native expression [59]
NMDAR Glun2b subunit GRIN2b Co-IP Native σ1 receptor Native GluN2a Rat hippocampus P2 pellet Native expression [59]
Human ether-a-go-go channel (hERG) KCNH2 Co-IP σ1 receptor-Myc Wildtype hERG HEK 293 cells Transient overexpression (σ1 receptor) [60]
Stable overexpression (hERG)
Anti-Myc chromatography myc-σ1 receptor hERG with HA tag between residues 443–444 tsA 201 cells Transient overexpression [58]
Proximity ligation myc-σ1 receptor hERG with HA tag between residues 443–444 tsA 201 cells Transient overexpression
HTRF myc-σ1 recepotr-HALO hERG with HA tag between residues 443–444 HEK 293 cells Transient overexpression
SK3 channel KCNN3 Co-IP σ1 receptor-Myc Wildtype SK3 SKmel28 cells Transient overexpression (σ1 receptor) [109]
Stable overexpression (SK3)
HTRF HALO-σ1 receptor-Myc SK3-HA HEK 293 cells Transient overexpression
Voltage-dependent N-type calcium channel (Cav2.2) CACNA1B FRET σ1 receptor-dsred EGFP-Cav2.2 HEK 293T cells Transient overexpression [51]
Co-IP σ1 receptor-dsred EGFP-Cav2.2 HEK 293T cells Transient overexpression
Voltage-dependent anion channel 2 (VDAC2) VDAC2 Co-IP Native σ1 receptor Native VDAC2 MA-10 cells Native expression [110]
Calcium release-activated calcium channel protein 1 ORAI1 Co-IP σ1 receptor-FLAG ORAI1-Myc tsA 201 cells Transient overexpression [39]
G-protein coupled Receptors (GPCRs)
μ opioid receptor (μ OR) OPRM1 Co-IP σ1 receptor-HA FLAG-μ OR HEK 293T cells Transient overexpression [40]
Pull down σ1 receptor-TEV μ OR (res.286–398) Purified protein Bacterial expression and purification [105]
D1 dopamine receptor (D1R) DRD1 BRET σ1 receptor-YFP D1R-Rluc HEK 293T Transient overexpression [41]
BRET σ1 receptor-Rluc D1R-YFP HEK 293T Transient overexpression
BRET σ1 receptor-Rluc YFP-D1R HEK 293T Transient overexpression
SRET σ1 receptor-YFP D1R-GFP HEK293T Transient overexpression [42]
Co-IP Native σ1 receptor Native D1R Mouse striatal slices Native expression
Proximity ligation Native σ1 receptor Native D1R Mouse striatal slices Native expression
D2 dopamine receptor (D2R) DRD2 BRET σ1 receptor-YFP D2R-Rluc HEK 293T Transient overexpression [43]
Proximity ligation Native σ1 receptor Native D2R Rat brain sections Native expression [44]
Cannabinoid receptor 1 (CB1) CNR1 Co-IP Native σ1 receptor Native CB1 Mouse brain synaptosomes Native expression [108]
BiFC σ1 receptor-split Venus CB1 split-Venus CHO cells Transient overexpression
Ghrelin receptor 1a (GHSR1a) GHSR BRET σ1 receptor-YFP GHSR1a-Rluc HEK 293T Transient overexpression [45]
Proximity ligation Native σ1 receptor Native GHSR1a Primary rat striatal neurons Native expression
Other proteins
Ankyrin B ANK2 Co-IP Native σ1 receptor Native ankyrin B NG-108 cells Native expression [33]
Co-IP Wildtype σ1 receptor Native ankyrin B MCF-7 cells Stable overexpression (σ1 receptor)
Native expression (ankyrin B)
[77]
Co-IP σ1 receptor (res. 102–223) Native ankyrin B MCF-7 cells Stable overexpression (σ1 receptor)
Native expression (ankyrin B)
Binding immunoglobulin protein (BiP) GRP78 Co-IP with crosslinking σ1 receptor-YFP Native BiP CHO cells Stable overexpression (σ1 receptor) [21]
Native expression (BiP)
Pull down σ1 receptor (res.116–223) Unknown source of recombinant BiP1 Purified protein Bacterial expression and purification
NMR σ1 receptor (res.112–223) Human BiP (res.24–654) Purified protein Bacterial expression and purification [76]
Co-IP Native σ1 receptor Native BiP Isolated bovine brain mitochondria Native expression [100]
Co-IP with crosslinking σ1 receptor-YFP Native BiP Neuro2A cells Transient overexpression (σ1 receptor) [111]
Native expression (BiP)
Dopamine transporter (DAT) DAT Co-IP GST-σ1 receptor myc-DAT HEK 293 cells Transient overexpression [61]
Co-IP Wildtype σ1 receptor Wildtype DAT HEK 293 cells Transient overexpression
BRET σ1 receptor-Rluc venus-DAT HEK 293 cells Transient overexpression
PD-L1 CD274 Co-IP σ1 receptor-HA PD-L1-FLAG MDA-MB-231 cells Transient overexpression [62]
Cerebroside synthase (UGT8) UGT8 Co-IP with crosslinking σ1 receptor-V5 UGT8-Myc CHO cells Transient overexpression [64]
IRE1 ERN1 Co-IP σ1 receptor-V5 Native IRE1 CHO cells Transient overexpression [63]
Pull down σ1 receptor (res.116–223) IRE1 (res.19–443)-V5-His Purified protein Bacterial expression and purification
TrkB NTRK2 Co-IP σ1 receptor-Myc HA-TrkB HEK 293T cells Transient overexpression [112]
Co-IP Native σ1 receptor Native TrkB Mouse cerebellar granule neurons Native expression
IL-24 IL24 Co-IP Native σ1 receptor Wildtype IL-24 DU145 cells Native expression (σ1 receptor) [113]
Viral overexpression (IL-24)
Bcl2 BAD Co-IP Native σ1 receptor Native Bcl2 Isolated bovine brain mitochondria Native expression [100]
Rac1 GTPase RAC1 Co-IP Native σ1 receptor Native Rac1 GTPase Isolated bovine brain mitochondria Native expression [100]
HINT1 HINT1 Pull down σ1 receptor-GST Wildtype HINT1 Purified protein Bacterial expression and purification [105]
Znf179 RNF112 Co-IP σ1 receptor-His Native Znf179 Neuro2A cells Transient overexpression (σ1 receptor) [114]
Native expression (Znf179)
Insig INSIG1 Co-IP σ1 receptor-FLAG Insig-Myc CHO cells Transient overexpression [64]
ELMOD1 ELMOD1 Co-IP FLAG-σ1 receptor GST-ELMOD1 HEK 293T cells Transient overexpression [115]
ELMOD2 ELMOD2 Co-IP FLAG-σ1 receptor GST-ELMOD1 HEK 293T cells Transient overexpression [115]
ELMOD3 ELMOD3 Co-IP FLAG-σ1 receptor GST-ELMOD1 HEK 293T cells Transient overexpression [115]
Steroidogenic acute regulatory protein (StAR) STAR Co-IP Native σ1 receptor Native StAR MA-10 cells Native expression [110]
Platelet derived growth factor β (PDGFβ ) PDGFRB Pull down Wildtype σ1 receptor GST-PDGFβ HEK 293T cells lysate (σ1 receptor) Transient overexpression (σ1 receptor) [116]
Purified protein (PDGFβ) Insect cell expression and purification (PDGFβ)
FRET σ1 receptor-RFP PDGFβ-GFP CHO cells Transient overexpression
Integrin β1 IGTB1 Co-IP Native σ1 receptor Native Integrin β1 MDA-MB-231 cells Native expression [117]
Emerin EMD Co-IP with crosslinking σ1 receptor-YFP Native Emerin Neuro2A cells Transient overexpression (σ1 receptor) [111]
Native expression (Emerin)
Co-IP with crosslinking σ1 receptor-V5-His Native Emerin Neuro2A cells Transient overexpression (σ1 receptor)
Native expression (Emerin)
Co-IP Native σ1 receptor Native Emerin Rat nucleus accumbens tissue Native expression
Co-IP Native σ1 receptor Native Emerin Mouse prefrontal cortex tissue Native expression
Lamin A/C LMNA Co-IP with crosslinking σ1 receptor-YFP Native Lamin A/C Neuro2A cells Transient overexpression (σ1 receptor) [111]
Native expression (Lamin A/C)
Co-IP with crosslinking σ1 receptor-V5-His Native Lamin A/C Neuro2A cells Transient overexpression (σ1 receptor)
Native expression (Lamin A/C)
Histone deacetylase 1 (HDAC1) HDAC1 Co-IP Native σ1 receptor Native HDAC1 Neuro2A cells Native expression [111]
Co-IP Native σ1 receptor Native HDAC1 Mouse prefrontal cortex tissue Native expression
Co-IP Native σ1 receptor Native HDAC1 NG-108 cells Native expression
Histone deacetylase 2 (HDAC2) HDAC2 Co-IP Native σ1 receptor Native HDAC2 Neuro2A cells Native expression [111]
Co-IP Native σ1 receptor Native HDAC2 Mouse prefrontal cortex tissue Native expression
Co-IP Native σ1 receptor Native HDAC2 NG-108 cells Native expression
Histone deacetylase 3 (HDAC3) HDAC3 Co-IP Native σ1 receptor Native HDAC3 Neuro2A cells Native expression [111]
Co-IP σ1 receptor-V5-His Native HDAC3 Neuro2A cells Transient overexpression (σ1 receptor)
Native expression (HDAC3)
Barrier-to autointegration-factor (BAF) BANF1 Co-IP Native σ1 receptor Native BAF Neuro2A cells Native expression [111]
Stromal interaction molecule 1 STIM1 Co-IP σ1 receptor-FLAG HA-STIM1 tSA 201 cells Transient overexpression [39]
1

The source of the recombinant BiP used in this paper is not stated.

While the modulation of ion channel [31, 3439] and GPCR [4045] signaling by σ1 receptor ligands is relatively well established, more work is needed to determine if these modulatory effects result from direct σ1-protein interactions. Multiple reports have shown instances where σ ligands directly modulate ion channels independently of the σ1 receptor [4649], and many of the reported effects of σ ligands on ion channels require concentrations of 10 μM or more despite nanomolar affinity for the receptor [31, 5056]. Additionally, evidence for direct σ1-protein interactions have relied primarily on Co-IP, resonance energy transfer (RET), or proximity ligation experiments (Table 1). These methods demonstrate proximity but cannot distinguish between direct and indirect interactions. Additionally, while Co-IP experiments can be informative, Co-IPs between membrane proteins are prone to false positives due to incomplete membrane solubilization and protein aggregation. Thus, it is desirable to validate molecular interactions suggested by such experiments with methods that can demonstrate direct molecular interactions. Ideally, these interactions are cross-validated with reconstituted biochemical or biophysical assays. Atomic force microscopy (AFM) has been used to investigate the properties of several σ1 receptor-ion channel complexes [36, 37, 57, 58], but this technique lacks sufficient resolution to show that the proteins are interacting in a specific manner with physiologically relevant affinities. Altogether, more work is needed to understand the mechanism of σ1 receptor modulation of GPCRs, ion channels, and other proteins.

The σ1 receptor as a ligand-operated chaperone

The identification of a large number of protein-protein interactions between the σ1 receptor and other proteins is consistent with the possibility that the σ1 receptor is a ligand-operated chaperone. This idea was first proposed by Hayashi and Su [21] and has since become a prevailing model in the field [2, 46]. In this model (Figure 2), the σ1 receptor exists in a resting state at the mitochondrion-associated membrane (MAM) of the endoplasmic reticulum (ER) [5]. While at the MAM, the σ1 receptor forms a complex with a chaperone called the binding immunoglobulin protein (BiP), which plays a central role in protein folding and quality control [5]. Activation of the σ1 receptor by small molecule agonists or by a decrease in ER calcium concentrations cause the σ1 receptor to dissociate from BiP and interact with client proteins in the ER or other organelles [2, 5] (Table 1; Figure 2).

Figure 2: A summary of the chaperone model for σ1 receptor function.

Figure 2:

The σ1 receptor has been proposed to act as a ligand-regulated chaperone to modulate multiple signaling pathways. Based on the text from Weng et al. [5]. Interaction partners are taken from the references in Table 1, and the localization of each partner was based on both the references in Table 1 and the Uniprot localization annotations for those proteins.

Though this model is widely accepted [2, 46], direct evidence for the receptor’s chaperone activity is relatively limited. To date, only one reconstituted biochemical experiment to demonstrate chaperone activity by the σ1 receptor has been published [21]. In this experiment, the authors monitored the ability of a purified C-terminal fragment of the σ1 receptor (residues 116–223; Figure 3A) to minimize the aggregation of proteins in a light scattering assay. While the C-terminal fragment of the receptor did reduce light scattering [21], recent structural work makes it unclear if this fragment fully recapitulates σ1 receptor function (see “Lessons from σ1 receptor crystal structures”).

Figure 3: The structure of the σ1 receptor.

Figure 3:

(A) the σ1 receptor’s amino acid sequence annotated by secondary structure, with α helices in blue and β sheets in orange. Histidine 116 is in red. (B) the structure of the human σ1 receptor (PDB ID: 5HK1). Each σ1 monomer is colored separately, and the membrane is represented by gray shading. The ligand PD 144418 is depicted in grey spheres. (C), (D), a σ1 receptor monomer, with amino acids 1–116 colored in orange (C) or hidden completely (D), to show parts of the protein that would remain if these residues were deleted. (E) a view of the ligand binding pocket (PDB ID: 5HK1). The red dashed line shows electrostatic interaction between the Glu172 and the basic nitrogen in the ligand. (F) Overlays of the structures of the σ1 receptor bound to the antagonist PD 144418 (PDB ID: 5HK1, blue) and the agonist (+)-pentazocine (PDB ID: 6DK1, orange). The red arrow shows the shift in helix α4 between the two structures. Waters unique to the (+)-pentazocine bound structure are depicted as red spheres. (G) The same overlay as in (F), but only helix α4 is colored and the rest of the receptor is shown in gray. Red arrows indicate the direction of the α4 shift induced by (+)-pentazocine.

Additional indirect evidence for the σ1 receptor’s chaperone function has been reported. For example, overexpression of the σ1 receptor can increase the whole-cell or surface expression of various proteins [38, 5962]. Similarly, siRNA knockdown of the σ1 receptor can decrease [21, 35, 60, 63] or increase [64] the expression of other proteins. Finally, σ1 overexpression or agonist treatment can protect cells against various forms of ER stress, while knockdown or antagonist treatment can make cells more vulnerable [21]. While these results are consistent with the idea that the σ1 receptor could be a chaperone protein, there are other possible explanations for this activity.

For example, while the σ1 receptor could be modulating signaling pathways through interactions with many proteins, it may also affect multiple pathways through only a subset of these interactions. The σ1 receptor has been shown to modulate the ER stress response and subsequent unfolded protein response (UPR), which can influence protein stability and localization [65, 66], presumably through binding and modulating the ER stress response regulatory protein IRE1 [63, 67]. A recent study demonstrated that the σ1 receptor regulates IRE1 activity in vivo, with σ1 receptor knockout enhancing IRE1 activation and the resulting inflammatory response in lipopolysaccharide-induced inflammation models [67]. Additionally, previous work suggests that the σ1 receptor is localized to cholesterol-rich lipid microdomains [68], where it can influence the distribution of lipids in the ER [68]. Changes in ER lipid composition are strongly associated with the ER stress response and the UPR [66]. By serving as a regulator of ER stress, the σ1 receptor could influence the signaling of many cellular pathways without needing to physically associate with more than a small number of proteins. Therefore, while the chaperone model of σ1 receptor function may be accurate, there is not yet sufficient data to rule out alternative models of σ1 receptor function.

σ1 receptor oligomerization

The most well-validated σ1 receptor protein-protein interaction is its association with itself. The formation of functional σ1 receptor oligomers was first suggested by BRET experiments performed in HEK 293T cells [43]. This was confirmed with a careful biochemical analysis, which showed that purified σ1 receptor existed in at least two different oligomeric states [69]. Later work in cells using both resonance energy transfer techniques [61, 70, 71] and native gels [61] has shown that the σ1 receptor exists in multiple oligomeric states and that ligands alter the distribution of these states. Antagonists bias the receptor towards higher molecular weight states, while agonists bias the receptor towards lower molecular weight states [61, 70, 71]. The precise functional consequences of σ1 receptor oligomerization remain to be determined.

Lessons from σ1 receptor crystal structures

In the last three years, crystal structures of the σ1 receptor have been solved in complex with five different ligands: PD 144418, haloperidol, NE-100, 4-IBP, and (+)-pentazocine [72, 73]. The receptor has five α helices including one transmembrane domain, and ten β strands, which make up the ligand binding domain (Figures 3A, 3B). Helices α4 and α5 are amphipathic helices that are partially embedded in the membrane. In all structures the receptor has crystallized as a homotrimer with an extensive inter-subunit interface (Figure 3B).

These structures have recast our perception of this protein’s fundamental architecture. First, these crystal structures have definitively established the σ1 receptor as a single-pass transmembrane protein (Figure 3B). Prior to this work, both single-pass and two-pass transmembrane models had been proposed for the σ1 receptor, though the two-pass model was most often discussed [34, 74]. However, crystal structures show that the receptor has a single transmembrane domain spanning residues 9–30 [72], and later proximity labeling [75] and BRET [71] experiments have corroborated these findings in cells.

Prior to the first reported crystal structures of the σ1 receptor, a nuclear magnetic resonance (NMR) study investigated the location of a putative second transmembrane domain of the receptor using a construct in which the first transmembrane domain had been removed [74]. In this study, the authors titrated lipid into their detergent mixture, and identified residues in the protein whose chemical shift values changed upon the addition of lipid [74]. The region comprising residues 91–107 was most sensitive to lipid titration and was tentatively identified as a second transmembrane domain based on the a priori assumption that such a domain exists [74]. However, in the crystal structure residues 91–107 form a buried hydrophobic β hairpin. Given the high hydrophobicity of this region it is not entirely surprising that it interacts with lipid, and this effect (rather than the existence of a second transmembrane domain) likely accounts for the results of this experiment.

As in the NMR studies, many other investigations have relied on the assumption of two transmembrane architecture to guide experimental design. These studies have often employed constructs lacking the N-terminal half of the protein based on the two-transmembrane model [21, 63, 76, 77]. While early work suggested that this construct may share some cellular functions with the native receptor [21, 77], the crystal structure shows that this truncation would remove the protein’s first three α helices and first four β strands, which make up about half of the ligand binding domain (Figures 3A, 3C and 3D). This brings the stability and functionality of this construct into question. Thus, it can be difficult to interpret work done using constructs based on the previous models of the σ1 receptor.

The recent structures reveal how the σ1 receptor is able to bind many structurally diverse ligands with high affinity [1]. The σ1 receptor interacts with most of its ligands through only a single electrostatic interaction between Glu172 and the basic nitrogen present in most σ1 receptor ligands (Figure 3E). In all five existing crystal structures, the rest of the ligand is free to fit into the large β-barrel-like binding pocket, which is lined with hydrophobic residues. Thus, as long as the ligand is chemically and sterically suited to the ligand binding pocket’s hydrophobicity and able to make the electrostatic interaction with Glu172, then the ligand may bind with high affinity. It should be noted that the σ1 receptor has also been shown to bind some neurosteroids, which do not have a basic nitrogen atom [1]. Presumably, these ligands would interact with the receptor differently, perhaps under conditions where Glu172 is protonated. However, the relatively modest affinity of neurosteroids for the σ1 receptor (200 nM or weaker) [1] has thus far prevented crystallization of a σ1-steroid complex.

With the crystal structure of the σ1 receptor bound to (+)-pentazocine [73], we also have the first glimpse as to how agonists and antagonists may differ at the structural level. The agonist (+)-pentazocine binds the receptor in the same binding pocket as antagonists, but it occupies a distinct region of this pocket from the four antagonist ligands co-crystallized with the receptor. This seems to induce a small conformational change in helix α4 that could explain why these ligands may bias the receptor towards smaller molecular weight states (Figures 3F, 3G). More work is required to see if this conformational change is caused by all σ1 receptor agonists, and to confirm if it indeed underlies the regulation of σ1 oligomerization.

Human genetics of the σ1 receptor

Ten pathogenic mutations in the human σ1 receptor gene have been reported in cohort studies (Table 2). In general, these appear to be loss of function mutations, resulting in either a form of juvenile-onset ALS known as ALS16 [7880], a form of distal hereditary motor neuropathy (dHMN) [8184], or other similar motor neuron deficits such as frontotemporal lobar degeneration with motor neuron disease (FTLD-MND) [85] or Silver syndrome (SS) [83]. These conditions feature gradual loss of motor neuron function, typically beginning in early childhood or adolescence. Mutations have been discovered in all four SIGMAR1 exons, as well as the 3’ untranslated region (3’ UTR) (Table 2). Though the molecular mechanism of pathogenicity is not known for all of these mutations, those mutants that have been studied often exhibit misfolding or mislocalization of the protein, resulting in cellular pathologies in the ER (Table 2).

Table 2: List of pathogenic human σ1 receptor mutations and their cellular effects.

A list of published mutations in the human SIGMAR1 gene that exhibit disease phenotypes. Only mutations reported in cohort studies are shown.

Variant Location on gene Amino acid change Phenotype Cellular effect
c.151+1G>T Exon 1 splice site Δ31–50 dHMN [81] Mislocalization [118]
c.194T>A Exon 2 L65Q dHMN/SS [83] Unknown
c.283dupC Exon 2 L95P + frameshift ALS [79, 119] Aberrant ER morphology [119]
c.304G>C Exon 2 E102Q ALS [78] Misfolding, ER stress [120], mislocalization [118]
c.412G>C Exon 3 E138Q dHMN [82] Mislocalization, aberrant ER function [82]
c.448G>A Exon 4 E150K dHMN [82] Mislocalization, aberrant ER function [82]
c.561_576del Exon 4 Stop codon after H69 dHMN [84] Unknown
Exon 4 deletion Exon Deletion of residues 69–223 dHMN [84] Unknown
c.672*31A>G
3’ UTR None ALS [80] Unknown
c.672*51G>T 3’ UTR None FTLD-MND [85] Increased mRNA expression [85]

σ1 receptor genetics and protein structure

The recent structures of the σ1 receptor offer an opportunity to structurally interpret the σ1 receptor mutations reported to cause human disease. Of the ten reported pathogenic mutations, four of them delete large sections of the receptor or introduce a premature frameshift or stop codon, and two are mutations in the SIGMAR1 gene’s 3’ UTR (Table 2). The other four mutations each substitute a single amino acid in the mature protein (Table 2). These pathogenic mutations are L65Q, E102Q, E138Q, and E150K (Table 2, Figure 4).

Figure 4: Structural locations of σ1 receptor disease mutations.

Figure 4:

The crystal structure of the σ1 receptor with amino acids L65, E102, E138, and E150 of chain A shown in orange, while the rest of chain A is shown in grey. Chains B and C are shown in blue and green, respectively. Dashed red lines represent hydrogen bonds. PD 144418 is shown in cyan. Waters are depicted as red spheres. (A), L65 is located on helix α4 and is surrounded by hydrophobic amino acids. Mutation to Q would likely be energetically unfavorable. (B), E102 makes two hydrogen bonds with backbone amide nitrogen atoms. Mutation to Q would replace one of these attractive bonds with a repulsive interaction, presumably destabilizing the protein. (C), E138 coordinates a complex network of water molecules and amino acids at the oligomeric interface. Mutation to Q would disrupt this network. (D), E150 makes a hydrogen bond with a backbone amide nitrogen to stabilize the β hairpin at the base of the ligand binding pocket’s “lid”. Mutation to K would prevent this interaction.

The σ1 receptor crystal structure provides a molecular rationale as to why these mutations could result in a nonfunctional receptor. The L65Q substitution would introduce a hydrophilic headgroup in a hydrophobic region of the receptor, which would be energetically unfavorable (Figure 4A). The substitutions E102Q, E138Q, and E150K would disrupt either intramolecular hydrogen bonds presumably necessary for proper folding of the receptor (E102Q and E150K, Figures 4B and 4D), or a hydrogen bonding network at the receptor’s oligomeric interface (E138Q, Figure 4C).

The σ2 receptor

In contrast to the σ1 receptor, relatively much less is known regarding the biological roles of the σ2 receptor. The σ2 receptor was discovered in 1990 through pharmacological profiling of cancer cell lines, and was defined as a binding site with high affinity for DTG and haloperidol but not benzomorphans [18]. Since then, the receptor has attracted considerable interest as a therapeutic target for the treatment of cancer [9, 11] and neurologic disease [15, 16]. Pharmacological experiments showed that the σ2 receptor is a 18 – 22 kDa intracellular membrane protein [18], and genetic knockout of the σ1 receptor revealed that the σ2 receptor was derived from a completely different gene than σ1. However, the gene that codes for the σ2 receptor was not known until very recently [86] (see “Molecular cloning of the σ2 receptor”). This was a major impediment to studying the biological function of the σ2 receptor, and as a result our understanding of its function is limited relative to that of other pharmacologically characterized receptors.

Molecular cloning of the σ2 receptor

The σ1 receptor was cloned in 1996 [28], but the gene that codes for the σ2 receptor eluded discovery despite multiple attempts to identify it [87, 88]. The most prominent attempt suggested that the σ2 receptor may be identical to the membrane-associated progesterone receptor membrane component 1 (PGRMC1) [88], but later work demonstrated that respective siRNA knockdown [89] and CRISPR knockout [90] of the murine and human PGRMC1 genes have no effect on σ2 binding in cells.

The σ2 receptor was finally identified as TMEM97 in 2017 [86]. This was determined via a an affinity chromatography approach in which a σ2-specific ligand fixed to a column was used to isolate candidate proteins from calf liver [86]. Candidates were identified by mass spectrometry and screened through heterologous expression and pharmacological profiling [86]. Expression of TMEM97 in cells lacking σ2 receptor confers a σ2 receptor binding profile to those cells, and siRNA knockdown of TMEM97 proportionally reduces σ2 binding [86], confirming that TMEM97 and the σ2 receptor are one and the same.

TMEM97/σ2 receptor biology and therapeutic potential

Currently, little is known about TMEM97 except that it resides in the endoplasmic reticulum and lysosomes [91], where it may bind to cholesterol [91] and regulate the Niemann-Pick protein NPC1 [92]. It is also overexpressed in some cancers [9395], which had also been reported for σ2 receptor before its identification [9].

Interest in the σ2 receptor/TMEM97 has centered around its role as a potential therapeutic target for the diagnosis and treatment of cancer [9], as well as the treatment of schizophrenia [15] and Alzheimer’s disease [16]. However, the lack of a gene for σ2 has prevented research that could unambiguously determine if the observed effects of σ2 receptor ligands are truly σ2-mediated, as it was impossible to knock down or overexpress the receptor. This is poised to change now that the receptor has been cloned. Already, one report has shown that some ligands that were thought to kill cancer cells through σ2 receptor in fact work through a σ2-independent mechanism [96].

Evolutionary connection between the σ1 and σ2 receptors

The σ1 receptor and the σ2 receptor/TMEM97 are not genetically related to one another, but they are both related to enzymes that perform the same function. The σ1 receptor’s closest relative is the yeast C8-C7 sterol isomerase ERG2p [28]. Similarly, the σ2 receptor/TMEM97 is related to emopamil binding protein (EBP), which is the mammalian C8-C7 sterol isomerase [97]. EBP and σ2 receptor/TMEM97 belong to the Expanded EBP superfamily (EXPERA), a small group of 5 proteins also including transmembrane 6 superfamily members 1 and 2 (TM6SF1 and TM6SF2), and Emopamil binding protein-like (EPBL) [98]. Thus, though σ1 and σ2 receptors are not genetically related, their similar pharmacological profiles are likely a consequence of convergent evolution.

Indeed, despite the fact that σ1 receptor and ERG2p are genetically dissimilar to EBP, all three proteins share similar pharmacological profiles [99]. Moebius et al. performed a detailed analysis comparing the pharmacological profiles of guinea pig σ1 receptor, ERG2p, and EBP from guinea pig, human, and mouse [99]. They found that all three proteins could bind several σ ligands with high affinity [99]. This raises the possibility that other EXPERA domain proteins may be tractable pharmacological targets. Currently, the functions of the σ2 receptor/TMEM97, TM6SF1, TM6SF2, and EBPL are not well understood, but it is possible that some or all of these proteins contribute to the physiological and behavioral effects reported for σ ligands.

Concluding remarks and future perspectives

The last five years have witnessed significant advances in our understanding of σ receptors. Crystal structures of the σ1 receptor [72, 73] provide a rationale for the receptor’s pharmacology and facilitate precise design of mutants and truncations for functional studies [27, 45, 61, 75]. Similarly, the identification of the σ2 receptor as TMEM97 will enable the use of modern molecular biological techniques in its study. However, a great deal of work remains if we are to understand even the basic biology of σ receptors (see Outstanding questions). Moving forward, σ receptor research must build on what has been done over the last 40 years while simultaneously assessing past work with a critical eye. Prevailing ideas should be revisited with newly developed tools to provide validation and mechanistic detail that is currently unavailable. New technologies such as CRISPR-Cas9 gene editing will help to clearly define which cellular effects of σ ligands are directly mediated by σ receptors. Though much remains to be done, this is an exciting time in σ receptor research, as our understanding of both receptors enters the molecular era.

Acknowledgements

We would like to thank Megan Sjodt and Meredith Skiba for a thoughtful reading and critique of the manuscript. This work was supported by a Klingenstein-Simons Fellowship in Neuroscience (A.C.K.), National Institutes of Health grant R01GM119185 (A.C.K.), the Winthrop Fund/Harvard Brain Science Initiative (A.C.K.) and National Science Foundation Graduate Research Fellowship award number DGE1745303 (H.R.S.).

Glossary

Agonist

A ligand that activates a receptor to elicit a biological signaling response

Antagonist

A ligand that inactivates a receptor to prevent or attenuate a biological signaling response

Atomic force microscopy (AFM)

A form of scanning probe microscopy that uses a physical probe to scan a surface, providing an image with sub-nanometer resolution

C8-C7 sterol isomerase

An enzyme involved in the synthesis of cholesterol/ergosterol. It moves a double bond between carbons C9 and C8 to C8 and C7

Chaperone

A class of protein that assists in the folding of other proteins. Many are essential parts of the cell’s protein synthesis machinery

Co-immunoprecipitation (Co-IP)

A technique in which an antibody against a “bait” protein attached to beads is used to remove the bait, and any proteins associated with it, from solution

CRISPR-Cas9

A gene editing system that uses the enzyme Cas9 with an associated guide RNA molecule to make modifications to specific regions of an organism’s DNA

G protein-coupled receptor (GPCR)

A member of a diverse family of seven-pass transmembrane receptor that couples to G proteins to transmit a biological signal

Inositol triphosphate (IP3)

A small molecule second messenger that activates the IP3 receptor, triggering calcium release from the ER

ion channel

A transmembrane protein that, when activated, allows specific ions to flow along their concentration gradient from one side of a membrane to another

Mitochondrion-associated membrane (MAM)

A specialized region of the ER membrane that forms a contact with the mitochondrial membrane, thought to be important for the control of calcium homeostasis, lipid metabolism, and autophagy

Pharmacophore

The part of a chemical structure that is responsible its specific interactions

Proximity ligation

A technique used to show that two proteins are within close proximity to one another. Cells are stained with primary antibodies against the proteins of interest. Secondary antibodies with complementary oligos are then added to bind to the primary antibodies. If the secondary antibodies are in close proximity, the oligos will anneal. Enzymes are added to initiate rolling DNA synthesis

Resonance energy transfer (RET)

A class of techniques used to show that two light-sensitive molecules are in close proximity to one another. A light-sensitive molecule is excited at a wavelength specific to that molecule. The excited molecule emits a photon at a wavelength that will excite the other light-sensitive molecule if the two are within close spatial proximity

siRNA knockdown

An RNA interference method that uses a short interfering RNA (siRNA) of 20–25 bp to reduce expression of the target gene through the RISC pathway

Unfolded protein response (UPR)

A cellular stress response to misfolded proteins in the ER lumen. The UPR includes a complex signaling cascade to fold or remove the unfolded proteins, or to induce apoptosis

References

  • 1.Walker JM, et al. , Sigma receptors: biology and function. Pharmacol Rev, 1990. 42(4): p. 355–402. [PubMed] [Google Scholar]
  • 2.Chu UB and Ruoho AE, Biochemical Pharmacology of the Sigma-1 Receptor. Mol Pharmacol, 2016. 89(1): p. 142–53. [DOI] [PubMed] [Google Scholar]
  • 3.Romero L, Merlos M, and Vela JM, Antinociception by Sigma-1 Receptor Antagonists: Central and Peripheral Effects. Adv Pharmacol, 2016. 75: p. 179–215. [DOI] [PubMed] [Google Scholar]
  • 4.Nguyen L, et al. , Role of sigma-1 receptors in neurodegenerative diseases. J Pharmacol Sci, 2015. 127(1): p. 17–29. [DOI] [PubMed] [Google Scholar]
  • 5.Weng TY, Tsai SA, and Su TP, Roles of sigma-1 receptors on mitochondrial functions relevant to neurodegenerative diseases. J Biomed Sci, 2017. 24(1): p. 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Smith SB, et al. , Sigma 1 receptor: A novel therapeutic target in retinal disease. Prog Retin Eye Res, 2018. 67: p. 130–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Katz JL, et al. , A role for sigma receptors in stimulant self-administration and addiction. Behav Pharmacol, 2016. 27(2–3 Spec Issue): p. 100–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Skuza G, Ethanol withdrawal-induced depressive symptoms in animals and therapeutic potential of sigma1 receptor ligands. Pharmacol Rep, 2013. 65(6): p. 1681–7. [DOI] [PubMed] [Google Scholar]
  • 9.Georgiadis MO, et al. , Sigma Receptor (sigmaR) Ligands with Antiproliferative and Anticancer Activity. Molecules, 2017. 22(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kim FJ and Maher CM, Sigma1 Pharmacology in the Context of Cancer. Handb Exp Pharmacol, 2017. 244: p. 237–308. [DOI] [PubMed] [Google Scholar]
  • 11.van Waarde A, et al. , Potential applications for sigma receptor ligands in cancer diagnosis and therapy. Biochim Biophys Acta, 2015. 1848(10 Pt B): p. 2703–14. [DOI] [PubMed] [Google Scholar]
  • 12.Bruna J, et al. , Efficacy of a Novel Sigma-1 Receptor Antagonist for Oxaliplatin-Induced Neuropathy: A Randomized, Double-Blind, Placebo-Controlled Phase IIa Clinical Trial. Neurotherapeutics, 2018. 15(1): p. 178–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.An extension study of ANAVEX2–73 in patients with mild to moderate Alzheimer’s disease (report no. ) 2018, Anavex Life Sciences Corp. [Google Scholar]
  • 14.Urfer R, et al. , Phase II trial of the Sigma-1 receptor agonist cutamesine (SA4503) for recovery enhancement after acute ischemic stroke. Stroke, 2014. 45(11): p. 3304–10. [DOI] [PubMed] [Google Scholar]
  • 15.Davidson M, et al. , Efficacy and Safety of MIN-101: A 12-Week Randomized, Double-Blind, Placebo-Controlled Trial of a New Drug in Development for the Treatment of Negative Symptoms in Schizophrenia. Am J Psychiatry, 2017. 174(12): p. 1195–1202. [DOI] [PubMed] [Google Scholar]
  • 16.Grundman M, et al. , A phase 1 clinical trial of the sigma-2 receptor complex allosteric antagonist CT1812, a novel therapeutic candidate for Alzheimer’s disease. Alzheimers Dement (N Y), 2019. 5Representative σ receptor ligands and the central: p. 20–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bowen WD, de Costa BR, Hellewell SB, Walker JM, and Rice KC, [3H](+)-Pentazocine: A potent and highly selective benzomorphan-based probe for sigma-1 receptors. Mol. Neuropharmacol, 1993. 3: p. 117–126. [Google Scholar]
  • 18.Hellewell SB and Bowen WD, A sigma-like binding site in rat pheochromocytoma (PC12) cells: decreased affinity for (+)-benzomorphans and lower molecular weight suggest a different sigma receptor form from that of guinea pig brain. Brain Res, 1990. 527(2): p. 244–53. [DOI] [PubMed] [Google Scholar]
  • 19.Glennon RA, Pharmacophore identification for sigma-1 (sigma1) receptor binding: application of the “deconstruction-reconstruction-elaboration” approach. Mini Rev Med Chem, 2005. 5(10): p. 927–40. [DOI] [PubMed] [Google Scholar]
  • 20.Schwarz S, Pohl P, and Zhou GZ, Steroid binding at sigma-”opioid” receptors. Science, 1989. 246(4937): p. 1635–8. [DOI] [PubMed] [Google Scholar]
  • 21.Hayashi T and Su TP, Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell, 2007. 131(3): p. 596–610. [DOI] [PubMed] [Google Scholar]
  • 22.Phan VL, et al. , Modulation of steroidal levels by adrenalectomy/castration and inhibition of neurosteroid synthesis enzymes affect sigma1 receptor-mediated behaviour in mice. Eur J Neurosci, 1999. 11(7): p. 2385–96. [DOI] [PubMed] [Google Scholar]
  • 23.Fontanilla D, et al. , The hallucinogen N,N-dimethyltryptamine (DMT) is an endogenous sigma-1 receptor regulator. Science, 2009. 323(5916): p. 934–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Nichols DE, N,N-dimethyltryptamine and the pineal gland: Separating fact from myth. J Psychopharmacol, 2018. 32(1): p. 30–36. [DOI] [PubMed] [Google Scholar]
  • 25.Keiser MJ, et al. , Predicting new molecular targets for known drugs. Nature, 2009. 462(7270): p. 175–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ramachandran S, et al. , The sigma1 receptor interacts with N-alkyl amines and endogenous sphingolipids. Eur J Pharmacol, 2009. 609(1–3): p. 19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Brailoiu E, et al. , Choline Is an Intracellular Messenger Linking Extracellular Stimuli to IP3-Evoked Ca(2+) Signals through Sigma-1 Receptors. Cell Rep, 2019. 26(2): p. 330–337 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hanner M, et al. , Purification, molecular cloning, and expression of the mammalian sigma1-binding site. Proc Natl Acad Sci U S A, 1996. 93(15): p. 8072–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Paul IA, et al. , Sigma receptors modulate nicotinic receptor function in adrenal chromaffin cells. FASEB J, 1993. 7(12): p. 1171–8. [DOI] [PubMed] [Google Scholar]
  • 30.Hayashi T, et al. , Modulation by sigma ligands of intracellular free Ca++ mobilization by N-methyl-D-aspartate in primary culture of rat frontal cortical neurons. J Pharmacol Exp Ther, 1995. 275(1): p. 207–14. [PubMed] [Google Scholar]
  • 31.Hayashi T, Maurice T, and Su TP, Ca(2+) signaling via sigma(1)-receptors: novel regulatory mechanism affecting intracellular Ca(2+) concentration. J Pharmacol Exp Ther, 2000. 293(3): p. 788–98. [PubMed] [Google Scholar]
  • 32.Novakova M, et al. , Highly selective sigma receptor ligands elevate inositol 1,4,5-trisphosphate production in rat cardiac myocytes. Eur J Pharmacol, 1998. 353(2–3): p. 315–27. [DOI] [PubMed] [Google Scholar]
  • 33.Hayashi T and Su TP, Regulating ankyrin dynamics: Roles of sigma-1 receptors. Proc Natl Acad Sci U S A, 2001. 98(2): p. 491–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Aydar E, et al. , The sigma receptor as a ligand-regulated auxiliary potassium channel subunit. Neuron, 2002. 34(3): p. 399–410. [DOI] [PubMed] [Google Scholar]
  • 35.Aydar E, et al. , Sigma-1 receptors modulate neonatal Nav1.5 ion channels in breast cancer cell lines. Eur Biophys J, 2016. 45(7): p. 671–683. [DOI] [PubMed] [Google Scholar]
  • 36.Balasuriya D, Stewart AP, and Edwardson JM, The sigma-1 receptor interacts directly with GluN1 but not GluN2A in the GluN1/GluN2A NMDA receptor. J Neurosci, 2013. 33(46): p. 18219–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Balasuriya D, et al. , The sigma-1 receptor binds to the Nav1.5 voltage-gated Na+ channel with 4-fold symmetry. J Biol Chem, 2012. 287(44): p. 37021–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kourrich S, et al. , Dynamic interaction between sigma-1 receptor and Kv1.2 shapes neuronal and behavioral responses to cocaine. Cell, 2013. 152(1–2): p. 236–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Srivats S, et al. , Sigma1 receptors inhibit store-operated Ca2+ entry by attenuating coupling of STIM1 to Orai1. J Cell Biol, 2016. 213(1): p. 65–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kim FJ, et al. , Sigma 1 receptor modulation of G-protein-coupled receptor signaling: potentiation of opioid transduction independent from receptor binding. Mol Pharmacol, 2010. 77(4): p. 695–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Navarro G, et al. , Direct involvement of sigma-1 receptors in the dopamine D1 receptor-mediated effects of cocaine. Proc Natl Acad Sci U S A, 2010. 107(43): p. 18676–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Moreno E, et al. , Cocaine disrupts histamine H3 receptor modulation of dopamine D1 receptor signaling: sigma1-D1-H3 receptor complexes as key targets for reducing cocaine’s effects. J Neurosci, 2014. 34(10): p. 3545–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Navarro G, et al. , Cocaine inhibits dopamine D2 receptor signaling via sigma-1-D2 receptor heteromers. PLoS One, 2013. 8(4): p. e61245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Borroto-Escuela DO, et al. , Cocaine self-administration specifically increases A2AR-D2R and D2R-sigma1R heteroreceptor complexes in the rat nucleus accumbens shell. Relevance for cocaine use disorder. Pharmacol Biochem Behav, 2017. 155: p. 24–31. [DOI] [PubMed] [Google Scholar]
  • 45.Aguinaga D, et al. , Cocaine Blocks Effects of Hunger Hormone, Ghrelin, Via Interaction with Neuronal Sigma-1 Receptors. Mol Neurobiol, 2018. [DOI] [PubMed] [Google Scholar]
  • 46.Amer MS, et al. , Inhibition of endothelial cell Ca(2)(+) entry and transient receptor potential channels by Sigma-1 receptor ligands. Br J Pharmacol, 2013. 168(6): p. 1445–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Gao XF, et al. , Sigma-1 receptor agonists directly inhibit Nav1.2/1.4 channels. PLoS One, 2012. 7(11): p. e49384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Brindley RL, et al. , Sigma-1 receptor ligands inhibit catecholamine secretion from adrenal chromaffin cells due to block of nicotinic acetylcholine receptors. J Neurochem, 2017. 143(2): p. 171–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Asano M, et al. , SKF-10047, a prototype Sigma-1 receptor agonist, augmented the membrane trafficking and uptake activity of the serotonin transporter and its C-terminus-deleted mutant via a Sigma-1 receptor-independent mechanism. J Pharmacol Sci, 2019. 139(1): p. 29–36. [DOI] [PubMed] [Google Scholar]
  • 50.Klette KL, et al. , Neuroprotective sigma ligands attenuate NMDA and trans-ACPD-induced calcium signaling in rat primary neurons. Brain Res, 1997. 756(1–2): p. 231–40. [DOI] [PubMed] [Google Scholar]
  • 51.Zhang K, et al. , Sigma-1 Receptor Plays a Negative Modulation on N-type Calcium Channel. Front Pharmacol, 2017. 8: p. 302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang H and Cuevas J, Sigma receptors inhibit high-voltage-activated calcium channels in rat sympathetic and parasympathetic neurons. J Neurophysiol, 2002. 87(6): p. 2867–79. [DOI] [PubMed] [Google Scholar]
  • 53.Katnik C, et al. , Sigma-1 receptor activation prevents intracellular calcium dysregulation in cortical neurons during in vitro ischemia. J Pharmacol Exp Ther, 2006. 319(3): p. 1355–65. [DOI] [PubMed] [Google Scholar]
  • 54.Renaudo A, et al. , Cancer cell cycle modulated by a functional coupling between sigma-1 receptors and Cl- channels. J Biol Chem, 2007. 282(4): p. 2259–67. [DOI] [PubMed] [Google Scholar]
  • 55.Herrera Y, et al. , sigma-1 receptor modulation of acid-sensing ion channel a (ASIC1a) and ASIC1a-induced Ca2+ influx in rat cortical neurons. J Pharmacol Exp Ther, 2008. 327(2): p. 491–502. [DOI] [PubMed] [Google Scholar]
  • 56.Johannessen M, et al. , Voltage-gated sodium channel modulation by sigma-receptors in cardiac myocytes and heterologous systems. Am J Physiol Cell Physiol, 2009. 296(5): p. C1049–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Carnally SM, et al. , Demonstration of a direct interaction between sigma-1 receptors and acid-sensing ion channels. Biophys J, 2010. 98(7): p. 1182–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Balasuriya D, et al. , A direct interaction between the sigma-1 receptor and the hERG voltage-gated K+ channel revealed by atomic force microscopy and homogeneous time-resolved fluorescence (HTRF(R)). J Biol Chem, 2014. 289(46): p. 32353–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Pabba M, et al. , NMDA receptors are upregulated and trafficked to the plasma membrane after sigma-1 receptor activation in the rat hippocampus. J Neurosci, 2014. 34(34): p. 11325–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Crottes D, et al. , Sig1R protein regulates hERG channel expression through a post-translational mechanism in leukemic cells. J Biol Chem, 2011. 286(32): p. 27947–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hong WC, et al. , The sigma-1 receptor modulates dopamine transporter conformation and cocaine binding and may thereby potentiate cocaine self-administration in rats. J Biol Chem, 2017. 292(27): p. 11250–11261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Maher CM, et al. , Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1. Mol Cancer Res, 2018. 16(2): p. 243–255. [DOI] [PubMed] [Google Scholar]
  • 63.Mori T, et al. , Sigma-1 receptor chaperone at the ER-mitochondrion interface mediates the mitochondrion-ER-nucleus signaling for cellular survival. PLoS One, 2013. 8(10): p. e76941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hayashi T, et al. , The lifetime of UDP-galactose:ceramide galactosyltransferase is controlled by a distinct endoplasmic reticulum-associated degradation (ERAD) regulated by sigma-1 receptor chaperones. J Biol Chem, 2012. 287(51): p. 43156–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Krebs J, Agellon LB, and Michalak M, Ca(2+) homeostasis and endoplasmic reticulum (ER) stress: An integrated view of calcium signaling. Biochem Biophys Res Commun, 2015. 460(1): p. 114–21. [DOI] [PubMed] [Google Scholar]
  • 66.Ho N, Xu C, and Thibault G, From the unfolded protein response to metabolic diseases - lipids under the spotlight. J Cell Sci, 2018. 131(3). [DOI] [PubMed] [Google Scholar]
  • 67.Rosen DA, et al. , Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Sci Transl Med, 2019. 11(478). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hayashi T and Su TP, Sigma-1 receptors (sigma(1) binding sites) form raft-like microdomains and target lipid droplets on the endoplasmic reticulum: roles in endoplasmic reticulum lipid compartmentalization and export. J Pharmacol Exp Ther, 2003. 306(2): p. 718–25. [DOI] [PubMed] [Google Scholar]
  • 69.Gromek KA, et al. , The oligomeric states of the purified sigma-1 receptor are stabilized by ligands. J Biol Chem, 2014. 289(29): p. 20333–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mishra AK, et al. , The sigma-1 receptors are present in monomeric and oligomeric forms in living cells in the presence and absence of ligands. Biochem J, 2015. 466(2): p. 263–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yano H, et al. , Pharmacological profiling of sigma 1 receptor ligands by novel receptor homomer assays. Neuropharmacology, 2018. 133: p. 264–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schmidt HR, et al. , Crystal structure of the human sigma1 receptor. Nature, 2016. 532(7600): p. 527–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Schmidt HR, et al. , Structural basis for sigma1 receptor ligand recognition. Nat Struct Mol Biol, 2018. 25(10): p. 981–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ortega-Roldan JL, et al. , Solution NMR studies reveal the location of the second transmembrane domain of the human sigma-1 receptor. FEBS Lett, 2015. 589(5): p. 659–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mavylutov T, et al. , APEX2- tagging of Sigma 1-receptor indicates subcellular protein topology with cytosolic N-terminus and ER luminal C-terminus. Protein Cell, 2018. 9(8): p. 733–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ortega-Roldan JL, Ossa F, and Schnell JR, Characterization of the human sigma-1 receptor chaperone domain structure and binding immunoglobulin protein (BiP) interactions. J Biol Chem, 2013. 288(29): p. 21448–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wu Z and Bowen WD, Role of sigma-1 receptor C-terminal segment in inositol 1,4,5-trisphosphate receptor activation: constitutive enhancement of calcium signaling in MCF-7 tumor cells. J Biol Chem, 2008. 283(42): p. 28198–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Al-Saif A, Al-Mohanna F, and Bohlega S, A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann Neurol, 2011. 70(6): p. 913–9. [DOI] [PubMed] [Google Scholar]
  • 79.Yang Y, et al. , Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med, 2013. 369(16): p. 1502–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ullah MI, et al. , In silico analysis of SIGMAR1 variant (rs4879809) segregating in a consanguineous Pakistani family showing amyotrophic lateral sclerosis without frontotemporal lobar dementia. Neurogenetics, 2015. 16(4): p. 299–306. [DOI] [PubMed] [Google Scholar]
  • 81.Li X, et al. , A SIGMAR1 splice-site mutation causes distal hereditary motor neuropathy. Neurology, 2015. 84(24): p. 2430–7. [DOI] [PubMed] [Google Scholar]
  • 82.Gregianin E, et al. , Loss-of-function mutations in the SIGMAR1 gene cause distal hereditary motor neuropathy by impairing ER-mitochondria tethering and Ca2+ signalling. Hum Mol Genet, 2016. 25(17): p. 3741–3753. [DOI] [PubMed] [Google Scholar]
  • 83.Horga A, et al. , SIGMAR1 mutation associated with autosomal recessive Silver-like syndrome. Neurology, 2016. 87(15): p. 1607–1612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Almendra L, et al. , SIGMAR1 gene mutation causing Distal Hereditary Motor Neuropathy in a Portuguese family. Acta Myol, 2018. 37(1): p. 2–4. [PMC free article] [PubMed] [Google Scholar]
  • 85.Luty AA, et al. , Sigma nonopioid intracellular receptor 1 mutations cause frontotemporal lobar degeneration-motor neuron disease. Ann Neurol, 2010. 68(5): p. 639–49. [DOI] [PubMed] [Google Scholar]
  • 86.Alon A, et al. , Identification of the gene that codes for the sigma2 receptor. Proc Natl Acad Sci U S A, 2017. 114(27): p. 7160–7165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Colabufo NA, et al. , Is the sigma2 receptor a histone binding protein? J Med Chem, 2006. 49(14): p. 4153–8. [DOI] [PubMed] [Google Scholar]
  • 88.Xu J, et al. , Identification of the PGRMC1 protein complex as the putative sigma-2 receptor binding site. Nat Commun, 2011. 2: p. 380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Abate C, et al. , Elements in support of the ‘non-identity’ of the PGRMC1 protein with the sigma2 receptor. Eur J Pharmacol, 2015. 758: p. 16–23. [DOI] [PubMed] [Google Scholar]
  • 90.Chu UB, et al. , The Sigma-2 Receptor and Progesterone Receptor Membrane Component 1 are Different Binding Sites Derived From Independent Genes. EBioMedicine, 2015. 2(11): p. 1806–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bartz F, et al. , Identification of cholesterol-regulating genes by targeted RNAi screening. Cell Metab, 2009. 10(1): p. 63–75. [DOI] [PubMed] [Google Scholar]
  • 92.Ebrahimi-Fakhari D, et al. , Reduction of TMEM97 increases NPC1 protein levels and restores cholesterol trafficking in Niemann-pick type C1 disease cells. Hum Mol Genet, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Han KY, et al. , Overexpression of MAC30 is associated with poor clinical outcome in human non-small-cell lung cancer. Tumour Biol, 2013. 34(2): p. 821–5. [DOI] [PubMed] [Google Scholar]
  • 94.Moparthi SB, et al. , Expression of MAC30 protein is related to survival and biological variables in primary and metastatic colorectal cancers. Int J Oncol, 2007. 30(1): p. 91–5. [PubMed] [Google Scholar]
  • 95.Ding H, et al. , Prognostic Value of MAC30 Expression in Human Pure Squamous Cell Carcinomas of the Lung. Asian Pac J Cancer Prev, 2016. 17(5): p. 2705–10. [PubMed] [Google Scholar]
  • 96.Zeng C, et al. , TMEM97 and PGRMC1 do not mediate sigma-2 ligand-induced cell death. Cell Death Discov, 2019. 5: p. 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Silve S, et al. , Emopamil-binding protein, a mammalian protein that binds a series of structurally diverse neuroprotective agents, exhibits delta8-delta7 sterol isomerase activity in yeast. J Biol Chem, 1996. 271(37): p. 22434–40. [DOI] [PubMed] [Google Scholar]
  • 98.Sanchez-Pulido L and Ponting CP, TM6SF2 and MAC30, new enzyme homologs in sterol metabolism and common metabolic disease. Front Genet, 2014. 5: p. 439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Moebius FF, et al. , Pharmacological analysis of sterol delta8-delta7 isomerase proteins with [3H]ifenprodil. Mol Pharmacol, 1998. 54(3): p. 591–8. [DOI] [PubMed] [Google Scholar]
  • 100.Natsvlishvili N, et al. , Sigma-1 receptor directly interacts with Rac1-GTPase in the brain mitochondria. BMC Biochem, 2015. 16: p. 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kubickova J, et al. , Haloperidol Affects Plasticity of Differentiated NG-108 Cells Through sigma1R/IP3R1 Complex. Cell Mol Neurobiol, 2018. 38(1): p. 181–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Tagashira H, Bhuiyan MS, and Fukunaga K, Diverse regulation of IP3 and ryanodine receptors by pentazocine through sigma1-receptor in cardiomyocytes. Am J Physiol Heart Circ Physiol, 2013. 305(8): p. H1201–12. [DOI] [PubMed] [Google Scholar]
  • 103.Kinoshita M, et al. , Sigma-1 receptor alters the kinetics of Kv1.3 voltage gated potassium channels but not the sensitivity to receptor ligands. Brain Res, 2012. 1452: p. 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Tchedre KT, et al. , Sigma-1 receptor regulation of voltage-gated calcium channels involves a direct interaction. Invest Ophthalmol Vis Sci, 2008. 49(11): p. 4993–5002. [DOI] [PubMed] [Google Scholar]
  • 105.Rodriguez-Munoz M, et al. , The ON:OFF switch, sigma1R-HINT1 protein, controls GPCR-NMDA receptor cross-regulation: implications in neurological disorders. Oncotarget, 2015. 6(34): p. 35458–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rodriguez-Munoz M, et al. , The sigma1 receptor engages the redox-regulated HINT1 protein to bring opioid analgesia under NMDA receptor negative control. Antioxid Redox Signal, 2015. 22(10): p. 799–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Rodriguez-Munoz M, Sanchez-Blazquez P, and Garzon J, Fenfluramine diminishes NMDA receptor-mediated seizures via its mixed activity at serotonin 5HT2A and type 1 sigma receptors. Oncotarget, 2018. 9(34): p. 23373–23389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Sanchez-Blazquez P, et al. , The calcium-sensitive Sigma-1 receptor prevents cannabinoids from provoking glutamate NMDA receptor hypofunction: implications in antinociception and psychotic diseases. Int J Neuropsychopharmacol, 2014. 17(12): p. 1943–55. [DOI] [PubMed] [Google Scholar]
  • 109.Gueguinou M, et al. , The SigmaR1 chaperone drives breast and colorectal cancer cell migration by tuning SK3-dependent Ca(2+) homeostasis. Oncogene, 2017. 36(25): p. 3640–3647. [DOI] [PubMed] [Google Scholar]
  • 110.Marriott KS, et al. , sigma-1 receptor at the mitochondrial-associated endoplasmic reticulum membrane is responsible for mitochondrial metabolic regulation. J Pharmacol Exp Ther, 2012. 343(3): p. 578–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tsai SY, et al. , Sigma-1 receptor mediates cocaine-induced transcriptional regulation by recruiting chromatin-remodeling factors at the nuclear envelope. Proc Natl Acad Sci U S A, 2015. 112(47): p. E6562–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kimura Y, et al. , Sigma-1 receptor enhances neurite elongation of cerebellar granule neurons via TrkB signaling. PLoS One, 2013. 8(10): p. e75760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Do W, et al. , Sigma 1 Receptor plays a prominent role in IL-24-induced cancer-specific apoptosis. Biochem Biophys Res Commun, 2013. 439(2): p. 215–20. [DOI] [PubMed] [Google Scholar]
  • 114.Su TC, et al. , The sigma-1 receptor-zinc finger protein 179 pathway protects against hydrogen peroxide-induced cell injury. Neuropharmacology, 2016. 105: p. 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ivanova AA, et al. , Characterization of recombinant ELMOD (cell engulfment and motility domain) proteins as GTPase-activating proteins (GAPs) for ARF family GTPases. J Biol Chem, 2014. 289(16): p. 11111–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Yao H, et al. , Cocaine hijacks sigma1 receptor to initiate induction of activated leukocyte cell adhesion molecule: implication for increased monocyte adhesion and migration in the CNS. J Neurosci, 2011. 31(16): p. 5942–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Palmer CP, et al. , Sigma-1 receptors bind cholesterol and remodel lipid rafts in breast cancer cell lines. Cancer Res, 2007. 67(23): p. 11166–75. [DOI] [PubMed] [Google Scholar]
  • 118.Wong AY, et al. , Aberrant Subcellular Dynamics of Sigma-1 Receptor Mutants Underlying Neuromuscular Diseases. Mol Pharmacol, 2016. 90(3): p. 238–53. [DOI] [PubMed] [Google Scholar]
  • 119.Watanabe S, et al. , Mitochondria-associated membrane collapse is a common pathomechanism in SIGMAR1- and SOD1-linked ALS. EMBO Mol Med, 2016. 8(12): p. 1421–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Dreser A, et al. , The ALS-linked E102Q mutation in Sigma receptor-1 leads to ER stress-mediated defects in protein homeostasis and dysregulation of RNA-binding proteins. Cell Death Differ, 2017. 24(10): p. 1655–1671. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES