11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Identification of the Sigma-2 Receptor: Distinct from the Progesterone Receptor Membrane Component 1 (PGRMC1)

      research-article
      Journal of alcoholism and drug dependence

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Editorial The sigma receptor (σR) subtypes, σ1 and σ2, have been mischaracterized [1,2]. A recent study suggested that the σ2R is the progesterone receptor membrane component 1 (PGRMC1) in rat livers. This finding was supported by the use of a novel photo affinity probe for σ2Rs, 5-[3-(4-[4azido2(4[6,7dimethoxy3,4dihydroisoquinolin 2(1H)yl]butylcarbamoyl)phenoxy]butyl)thioureido]-2-(6-hydroxy-3-oxo-3H-xanthen-9-l)benzoic acid (WC-21) [3]. Since that study, many have accepted that these two entities are the same. More recent studies have, however, indicated that this identification was mischaracterized [4,5]. This mischarecterization is significant for the establishment of σ2R pharmacology. Precise pharmacological characterization of the σ2R is important because it has been implicated with stimulant abuse [6,7]. σRs are unique intracellular chaperone proteins [8] initially thought to be opioid receptor subtypes [9]. They have been classified into two subtypes based on specific radioligand binding assays using [3H](+)-pentazocine for σ1Rs and [3H]1,3-di-o-tolylguanidine ([3H]DTG, in the presence of dextrallorphan to mask the σ1R) for σ2Rs in rat liver and kidney membranes [10]. Currently, the more selective σ1R ligand (+)-pentazocine has replaced dextrallorphan to mask the σ1R [7,11-14]. The σ1R has already been cloned as a 25-29 kDa chaperone protein composed of 223 amino acids [4,8,15]. It is widely distributed throughout the body [16-20]. Upon binding with agonists or under cellular stress, σ1Rs translocate from their primary endoplasmic reticulum (ER) location to different subcellular compartments where they can regulate ion channels and G-protein-coupled-receptor (GPCR) signaling [8,21-24]. In vivo functional studies on σ1Rs suggest that they play a substantial role in various cellular functions. Drugs acting at this receptor have been studied for their potential therapeutic effects in cancer, human immunodeficiency virus (HIV) infection, various psychiatric disorders, and substance abuse [1,25]. The σ1R is not a GPCR. Thus, it is challenging to determine what constitutes an agonist or an antagonist. For example, in vitro studies using NG-108 and Chinese Hamster Ovary (CHO) cells have demonstrated that the selective σ1R ligands PRE-084 and (+)pentazocine can dose-dependently cause the dissociation of σ1R from a binding immunoglobulin protein/78 kDa glucose-regulated protein (BiP/GRP-78), another ER chaperone [8,26]. Thus, they serve as agonists. In contrast, the σ1R ligands haloperidol and 4-methoxy-3-(2-phenylethoxy)-N,N-dipropylbenzeneethanamine (NE-100) alone do not affect the σ1R-BiP association but both completely inhibit the dissociation of σ1R from BiP caused by (+)pentazocine: they serve as antagonists [8,26]. In vivo, however, there is--as yet--no established functional assay for the σR subtypes. However, there is evidence showing a dose-dependent antagonism in vivo using the in vitro σ1R antagonists against the in vitro σ1R agonists using drug self-administration procedures [7,12,27,28]. Thus, it appears that the in vitro agonist-antagonist relationship will apply some in vivo responses. The [3H] (+)-pentazocine-inaccessible σR, the σ2R, is an 18-21 kDa protein that has not been cloned yet [3,20,29-31]. However, a previous study using the radioligands [3H](+)-pentazocine, and [3H]DTG (in the presence of dextrallorphan) and a Flotillin-2 dotblotting technique in rat liver membranes found that σ2Rs are primarily localized in membrane lipid rafts whereas the σ1R localization appears in both raft and non-raft membrane domains [32]. The σ1R is dynamic and can translocate from its primary ER location to different subcellular compartments [24]. Previous mass spectrometry studies identified the σ2R-like proteins as being dimers consisting of H2A/H2B, the human nucleosomal proteins [33,34], which were defined using [3H]1-cyclohexyl-4-[3-(5-methoxy-1,2,3,4-tetra-hydronaphthalen-1-yl)propyl]piperazine ([3H]PB28) as a radioligand having a 19-fold higher affinity for the σ2 than for the σ1 receptors [35]. Abate et al. [34] showed that [3H]PB28 accumulation was up to five-fold higher in nuclear fractions than in cytosolic fractions in SK-N-SH and MCF7 cells. However, the dimer differs from the σ2R in membrane association [32]. Thus, the identity of σ2Rs as nucleosomal proteins does not appear to be viable. Due to the lack of a known σ2R amino acid sequence, photoaffinity labeling remains the most viable approach for visualizing the receptor using sodium dodecyl sulfate (SDS) gels [29]. The basic principle is to covalently combine a photoactivatable σ2R-binding probe with the receptor such that the probe (radioactive- or fluorescent-labeled) remains with the protein even after denaturation with SDS [29]. Using a novel photoaffinity probe for σ2Rs, WC-21, a recent study identified the σ2R as the PGRMC1 in rat livers [3]. For example, the non-selective σ1/2R ligand DTG prevented the photolabeling of PGRMC1 (with WC-21) [3]. Further, an immunocytochemical study revealed that both PGRMC1 and (1R,3r,5S)-9-(10-[(7-Nitrobenzo[c] [1,2,5]oxadiazol-4-yl)amino]decyl)-9 azabicyclo[3.3.1]nonan-3-yl (2-methoxy-5-methylphenyl) carbamate (SW120), a fluorescent σ2R ligand, colocalize with molecular markers of the ER and mitochondria in HeLa cells [3]. As noted for the σ1R, studies utilizing various in vitro techniques indicated that σ2Rs are intracellular proteins. However, the affinity of DTG for the PGRMC1 was not reported in the study [3]. Nonetheless, it appears that the identification of the σ2R as the PGRMC1 [3] has been accepted widely. However, two recent studies [1,2] demonstrated a more viable data set against this identification as follows: The molecular size of PGRMC1 (25 kDa) is approximately 7 kDa higher than that of the σ2R (∼ 18 kDa) [4]. Using specific photolabeling with [125I]-iodoazido-fenpropimorph ([125I]-IAF), the photolabeled σ2R band was not diminished in NSC34 cells devoid of or overexpressing the PGRMC1 [4]. Further, PGRMC1 knockout did not reduce [125I]-IAF photolabeling of the σ2R (18-21 kDa band) that was protectable by DTG and the highly σ2R-selective CM compounds [e.g. 1-(4-[6,7-dimethoxy-3,4-dihydroisoquinolin-2(1H)-yl]butyl)-3-methyl-1H-benzo[d]imidazol-2(3H)-one hydrochloride (CM 398)] [4]. The lack of influence of PGRMC1 knockout on the photolabeling of σ2R indicates a lack of a σ2R ligand-binding pocket formed by PGRMC1/σ2R complexes. The results also suggest that the σ2R is not a splice variant of the PGRMC1, thus, these two proteins are derived from different genes. Alternatively, the PGRMC1 may be another DTG-binding protein that does not bind the photoprobe [125I]-IAF. If PGRMC1 is a high-affinity DTG binding site, elevation of PGRMC1 protein levels would result in an increase in maximal binding of [3H]DTG. However, neither the Bmax nor Kd values for [3H]DTG changed significantly in response to PGRMC1 overexpression, knockout or silencing in NSC34 cells [4] or human MCF7 adenocarcinoma cell lines [5] which are devoid of the σ1R [36]. Progesterone has been reported to be a high-affinity (Kd=35 nM) ligand for PGRMC1 (Table 1). However, the Ki value of progesterone for the σ2R [4] is approximately 406-fold higher than the Kd value for PGRMC1 in rat liver membranes (Table 1). Further, the Ki value of DTG for the PGRMC1 is 472,000 ± 420,000 nM (Table 1) using cold (+)-pentazocine to block the σ1R [4], which is approximately 15,000-fold higher than that for the σ2R [4] (Table 1). However, the Ki value of DTG for the PGRMC1 [4] was shown to be >1,000-fold lower than that obtained in a previous study [37] (Table 1). This discrepancy likely results from the lack of use of a selective cold blocker at the σ1R in the previous study [37] since DTG can also bind the σ1R with high affinity (Table 1). The binding profile of DTG for the PGRMC1 has been consistent with that for haloperidol, another non-selective σ1/2R ligand [4] (Table 1). Thus, the PGRMC1 is not a high-affinity DTG binding site, which also means that the PGRMC1 is not the σ2R. Together, these new data [4,5] clearly suggest that the σ2R and PGRMC1 are two different molecular entities. Furthermore, the photo affinity probe containing a σ2R-directing moiety that led to the identification of PGRMC1 [3] as the σ2R (with WC-21), likely binds both σ2R and PGRMC1. The identification of the σ2R as distinct from the PGRMC1 [4,5] should have considerable impact especially in the cancer study field since the σ2R has been developed as a biomarker for various tumor cells [38]. Other studies have attempted to examine the correlation between the binding affinity of various σR ligands and their ability to produce effects both in vitro and in vivo through the σ2R [35,39]. However, the evidence for σ2R-mediated actions from these studies is not compelling because of the mixed use of σR agonist-like and antagonist-like ligands. Thus, the pharmacology and physiological role of σ2Rs remain undetermined due to unsuccessful efforts to clone the receptor and a lack of selective ligands. On the other hand, in vitro functional studies have demonstrated that the activation of the σ2R resulted in the synthesis and release of dopamine in the rat brain [6,7]. Thus, future studies that further explore σ2R pharmacology may result in a better understanding of the dopamine-mediated reinforcing mechanism associated with stimulant abuse and other dopamine-related diseases (e.g. Parkinson's disease and schizophrenia).

          Related collections

          Most cited references40

          • Record: found
          • Abstract: found
          • Article: not found

          Purification, molecular cloning, and expression of the mammalian sigma1-binding site.

          Sigma-ligands comprise several chemically unrelated drugs such as haloperidol, pentazocine, and ditolylguanidine, which bind to a family of low molecular mass proteins in the endoplasmic reticulum. These so-called sigma-receptors are believed to mediate various pharmacological effects of sigma-ligands by as yet unknown mechanisms. Based on their opposite enantioselectivity for benzomorphans and different molecular masses, two subtypes are differentiated. We purified the sigma1-binding site as a single 30-kDa protein from guinea pig liver employing the benzomorphan(+)[3H]pentazocine and the arylazide (-)[3H]azidopamil as specific probes. The purified (+)[3H]pentazocine-binding protein retained its high affinity for haloperidol, pentazocine, and ditolylguanidine. Partial amino acid sequence obtained after trypsinolysis revealed no homology to known proteins. Radiation inactivation of the pentazocine-labeled sigma1-binding site yielded a molecular mass of 24 +/- 2 kDa. The corresponding cDNA was cloned using degenerate oligonucleotides and cDNA library screening. Its open reading frame encoded a 25.3-kDa protein with at least one putative transmembrane segment. The protein expressed in yeast cells transformed with the cDNA showed the pharmacological characteristics of the brain and liver sigma1-binding site. The deduced amino acid sequence was structurally unrelated to known mammalian proteins but it shared homology with fungal proteins involved in sterol synthesis. Northern blots showed high densities of the sigma1-binding site mRNA in sterol-producing tissues. This is also in agreement with the known ability of sigma1-binding sites to interact with steroids, such as progesterone.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The pharmacology of sigma-1 receptors.

            Originally considered an enigmatic protein, the sigma-1 receptor has recently been identified as a unique ligand-regulated molecular chaperone in the endoplasmic reticulum of cells. This discovery causes us to look back at the many proposed roles of this receptor, even before its molecular function was identified, in many diseases such as methamphetamine or cocaine addiction, amnesia, pain, depression, Alzheimer's disease, stroke, retinal neuroprotection, HIV infection, and cancer. In this review, we examine the reports that have clearly shown an agonist-antagonist relationship regarding sigma-1 receptors in models of those diseases and also review the relatively known mechanisms of action of sigma-1 receptors in an attempt to spur the speculation of readers on how the sigma-1 receptor at the endoplasmic reticulum might relate to so many diseases. We found that the most prominent action of sigma-1 receptors in biological systems including cell lines, primary cultures, and animals is the regulation and modulation of voltage-regulated and ligand-gated ion channels, including Ca(2+)-, K(+)-, Na(+), Cl(-), and SK channels, and NMDA and IP3 receptors. We found that the final output of the action of sigma-1 receptor agonists is to inhibit all above-mentioned voltage-gated ion channels, while they potentiate ligand-gated channels. The inhibition or potentiation induced by agonists is blocked by sigma-1 receptor antagonists. Other mechanisms of action of sigma-1 receptors, and to some extent those of sigma-2 receptors, were also considered. We conclude that the sigma-1 and sigma-2 receptors represent potential fruitful targets for therapeutic developments in combating many human diseases.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The effects of morphine- and nalorphine- like drugs in the nondependent and morphine-dependent chronic spinal dog.

              Three different syndromes produced by congeners of morphine have been identified in the nondependent chronic spinal dog. These syndromes have been attributed to interaction of agonists with three distinguishable receptors (mu, kappa and sigma). Morphine is the prototype agonist for the mu receptor, ketocyclazocine for the kappa receptor and SKF-10,047 for the sigma receptor. The morphine syndrome (mu) in the dog is characterized by miosis, bradycardia, hypothermia, a general depression of the nociceptive responses and indifference to environmental stimuli. Ketocyclazocine (kappa) constricts pupils, depresses the flexor reflex and produces sedation but does not markedly alter pulse rate or the skin twitch reflex. SKF-10,047 (sigma), in contrast to morphine and ketocyclazocine, causes mydriasis, tachypnea, tachycardia and mania. The effects of these three drugs can be antagonized by the pure antagonist naltrexone, indicating that they are agonists. Further, chronic administration of morphine, ketocyclazocine and SKF-10,047 induces tolerance to their agonistic effects. Morphine suppresses abstinence in morphine-dependent dogs while ketocyclazocine does not. Ketocyclazocine at best precipitated only a liminal abstinence syndrome in the morphine-dependent dog, indicating that it had little affinity for the morphine receptor. Ketocyclazocine thus appears to be a selective agonist at the kappa receptor. Further, it has been shown that buprenorphine is a partial agonist of the mu type which both suppressed and precipitated abstinence in the morphine-dependent dog while morphine and propoxyphene are stronger agonists. Apomorphine and SKF-10,047 produce similar pharmacologic effects suggesting that sigma activity may involve a dopaminergic mechanism.
                Bookmark

                Author and article information

                Contributors
                Journal
                101608872
                41412
                J Alcohol Drug Depend
                J Alcohol Drug Depend
                Journal of alcoholism and drug dependence
                2329-6488
                27 June 2016
                25 April 2016
                2016
                01 July 2016
                : 4
                : doi:10.4172/2329-6488.1000e130
                Affiliations
                Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), USA
                Author notes
                [* ]Corresponding author: Takato Hiranita, Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR 72079-9501, USA, Tel: 870-543-7937; Fax: 870-543-7745; takato.hiranita@ 123456fda.hhs.gov
                Article
                NIHMS798364
                10.4172/2329-6488.1000e130
                4930110
                27376101
                ae1377b7-73b7-4b3d-aa0c-06687170eaab

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                Categories
                Article

                Comments

                Comment on this article