Next Article in Journal
Pharmacokinetics and Ex Vivo Activity of 7-Methylxanthine, an Inhibitor of Monosodium Urate Crystallization
Previous Article in Journal
Pharmacological Evaluation of Araliadiol as a Novel Anti-Inflammatory Agent in LPS-Induced RAW 264.7 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological and Pathological Implications of Sigma-1 Receptor in Neurodegenerative Diseases

Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, P.O. Box 19626, Springfield, IL 62794-9626, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(6), 1409; https://doi.org/10.3390/biomedicines13061409
Submission received: 9 April 2025 / Revised: 29 May 2025 / Accepted: 6 June 2025 / Published: 8 June 2025
(This article belongs to the Special Issue Cell Signaling and Molecular Regulation in Neurodegenerative Disease)

Abstract

:
Originally identified as a potential receptor for opioids, the sigma-1 receptor is now recognized as an intracellular chaperone protein associated with mitochondria-associated membranes at the endoplasmic reticulum (ER). Over the past two decades, extensive research has revealed that the sigma-1 receptor regulates many cellular processes, such as calcium homeostasis, oxidative stress responses, protein folding, and mitochondrial function. The various functions of the sigma-1 receptor highlight its role as a central modulator of neuronal health and may be a promising pharmacological target across multiple neurodegenerative conditions. Herein, we provide an overview of the current pharmacological understanding of the sigma-1 receptor with an emphasis on the signaling mechanisms involved. We examine its pathological implications in common neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease, and multiple sclerosis. We then highlight how sigma-1 receptor modulation may influence disease progression as well as potential pharmacological mechanisms to alter disease outcomes. The translational potential of sigma-1 receptor therapies is discussed, as well as the most up-to-date results of ongoing clinical trials. This review aims to clarify the therapeutic potential of the sigma-1 receptor in neurodegeneration and guide future research in these diseases.

1. Introduction

Sigma receptors were first identified in the 1970s during investigations into the mechanisms of opioid action. W.R. Martin observed that SKF-10047, a derivative of morphine, caused mydriasis, tachypnea, tachycardia, and mania, distinct from those induced by known opioid receptor agonists such as morphine, a mu receptor agonist, and ketocyclazocine, a kappa receptor agonist [1]. These observations suggested that SKF-10047 was binding to an unrecognized receptor, as opposed to the traditional mu, kappa, and delta receptor subtypes. The “sigma” classification, derived from the Greek letter σ, is used to denote its distinct binding properties [1]. This discovery initiated considerable interest in identifying novel pathways through which the opioid-like compounds could exert their effects.
The 1980s and 1990s marked a turning point for sigma receptor research. Studies by Tam and Cook in 1984 demonstrated that the effects of sigma ligands could not be reversed by naloxone, an opioid antagonist, which is a defining feature of opioid receptors [2]. This observation led to the recognition that sigma receptors are pharmacologically distinct from opioid receptors due to their unique binding sites, affinities, pharmacological profile, and naloxone insensitivity [3].
Later, based on pharmacological profiles, binding characteristics, and tissue distribution patterns, sigma receptors were categorized into the sigma-1 receptor (S1R) and the sigma-2 receptor (S2R) [4]. For instance, S1R exhibits a high affinity for (+)-pentazocine, whereas S2R does not. In contrast, other ligands show preferential binding to S2R. These distinctions, paired with differences in molecular weight and other biochemical properties, laid the groundwork for distinguishing between the two receptor subtypes.
The cloning of S1R in 1996 by Hanner et al. marked a significant milestone, providing insight into its molecular structure and function [5]. The gene for S1R encodes a protein that differs from the traditional G protein-coupled receptors and instead shares some characteristics with molecular chaperones. In contrast, the molecular identity of the S2R remained elusive until 2017, when it was identified as a distinct entity from S1R and was shown to be involved in different cellular processes [6], representing another significant advancement in sigma receptor research.
Initially mistaken as a subtype of opioid receptors, sigma receptors have emerged as a distinct class of proteins with diverse roles in cellular signaling, neurophysiology, and pharmacology. Their discoveries have opened new avenues for understanding disease mechanisms and developing novel therapeutic strategies.
To address the growing interest in S1R-related therapeutics in neurodegeneration, we conducted a scoping review of the literature using a structured PubMed search. The review focused on articles published in the last decade (2015–2025). Terms that were searched included “sigma receptor,” “σ-1 receptor,” “σ-2 receptor,” among other related variations of the term. Emphasis was placed on mechanistic and translational studies.
Given the broad functions of S1R and its numerous implications in pathological states, the structure of the review is split into diseases. We begin by exploring the structure and biology and move forward to pathologies. Of the pathologies, mechanical insults to the nervous system are first discussed. We subsequently review the implications of several neurodegenerative diseases. Each section reviews the implications of S1R in the development of these various pathological states, how pharmaceuticals may modulate this process using S1R, and the most up-to-date research, ranging from animal models to clinical trials. A schematic of the review is represented in Figure 1. Additionally, we provide an updated review of the literature with the newest results in the past five years.

2. Sigma-1 Receptor: Structure, Functions, and Pharmacology

S1R is a novel protein localized to the endoplasmic reticulum (ER), where it associates with lipid rafts [1,7,8,9]. The first cloning took place in 1996 and localized the gene to chromosome 9p13 [5,10]. S1R was later resolved structurally as a homo-trimer with a unique transmembrane domain for each region [5,11,12,13,14,15]. Its oligomerization state is modulated by ligand binding, as agonists promote multimer formation, whereas antagonists reduce it [16]. S1R is additionally capable of forming heteromeric complexes with other receptors, such as the dopamine receptor [17,18]. With a molecular weight of 25 kDa, S1R is structurally unique and shares no homology to other known proteins in the genome [9,19].
Following the identification of S1R, S2R was described, though it remains less characterized. Like S1R, S2R binds to SKF-10047 and associates with lipid rafts [7,20,21,22]. Additional research has shown that S2R is potentially a binding site for progesterone receptor membrane component 1 (Pgrmc1) [23] and was later identified as the known transmembrane protein TMEM97 [6]. Currently, S1R and S2R are primarily distinguished based on ligand binding assays. H3(+) pentazocine has a selective affinity for S1R, whereas H3-DTG or H3-(+)-3-PPP combined with a masking S1R ligand, such as pentazocine, is used to selectively label S2R [24,25].
S1R plays many roles in cellular biology. It is capable of translocating between the ER, mitochondria, and plasma membrane [26,27,28,29]. In addition, S1R interacts with G-proteins, ion channels, inositol 1,4,5-triphosphate (IP3) receptors, and glutamate receptors, mediating diverse signaling pathways [29,30,31]. It is widely expressed in multiple vital organs, such as the heart, liver, and kidney, as well as in immune cells [32]. Pharmacologically, S1R binds a wide variety of ligands, such as antipsychotics, antidepressants, and neurosteroids [3,33]. Currently, it has been implicated in cell survival, as its expression is elevated in various cancers. Furthermore, S1R acts as a molecular chaperone with established roles in neuroprotection [8,34,35,36,37].
In the resting state, S1R is localized to mitochondria-associated ER membranes (MAM), where it resides in ceramide- and cholesterol-rich microdomains with the ER chaperone BiP [27,38]. Under times of cellular stress, ER dysfunction prompts S1R to dissociate from BiP. Upon dissociation, S1R binds IP3 receptors, enhancing cell survival through calcium signaling between the ER and mitochondria. Previous testing has shown that S1R agonists amplify this stress response while antagonists inhibit it [27]. An overview of the structure and function of S1R is represented in Figure 2.
Overall, under physiological conditions, S1R plays a minor role but transitions to an active chaperone protein in times of stress, supporting cell survival [30]. Aside from the IP3 pathway, S1R modulates dopaminergic and cholinergic transmission by influencing associated ion channels [39,40,41,42].

3. Sigma-1 Receptor in Nerve Injury

Nervous system injuries can result from a range of insults, including mechanical trauma and ischemia. These can affect both central and peripheral structures [43,44,45]. Following nerve injury, S1R expression has been shown to increase [46]. In ischemic stroke, S1R is hypothesized to exert neuroprotective effects by preventing neural apoptosis and inflammation while promoting neurotrophic signaling [47,48]. A primary driver of apoptosis following ischemic stroke includes endoplasmic reticulum stress, which can be attenuated by S1R activation with dexmedetomidine or aniline derivatives by reducing ER stress proteins CHOP, Caspase-3, and phosphorylated-JNK [48,49].
Strokes remain one of the leading causes of disability worldwide, with ischemic events composing the majority [50]. Generally, the prognosis of a stroke often correlates with the extent of ischemia, representing the amount of irreversibly damaged neural tissue [51]. Preclinical studies with the S1R agonists N,N-Dimethyltryptamine (DMT), and oxeladin have demonstrated potential in reducing the size of an infarct and improving long-term outcomes. DMT additionally reduced APAF1 levels, a marker of apoptosis, as well as inflammatory markers TNF-α, IL1-β, and IL-6, while increasing BDNF levels, central to neural preservation. Early-phase clinical trials for ischemic stroke in healthy individuals confirmed the safety of varying doses of DMT, opening avenues for future treatments [47,52,53,54]. Other markers of apoptosis, such as Bax and caspase-3, were markedly reduced in microglial cells following a stroke when treated with the S1R agonist afobazole [55]. Reactive gliosis following ischemic stroke is a maladaptive reaction that scars the brain parenchyma [56]. The S1R agonist afobazole, as well as the S2R antagonist S1RA, reduced the number of reactive cells through a reduction in matrix metalloproteinase-9, contributing to better recovery [55,57]. Aside from limiting acute injury, S1R activation through the selective agonist PRE-084 promoted recovery of white matter injury, decreased demyelination, and increased myelination factors CNPase, MOG, and PDGFRα after a stroke in preclinical models [58]. Following an ischemic stroke, macrophage-mediated efferocytosis is critical for clearing cellular debris from the infarct and inducing neural repair and inflammation resolution. Recent studies show that S1R knockout (KO) models impair this process and exacerbate brain damage following an ischemic stroke [59,60]. The benefits of S1R agonism following stroke are not limited to motor recovery but also suggest cognitive improvement in post-stroke models when treated with the S1R agonists ulinastatin, oxeladin, and PRE-084 by rescuing BDNF through the NR2A-CaMKIV-TORC1 pathway [52,61,62,63]. Neurite outgrowth is another central mechanism to post-stroke recovery, which was induced with the S1R agonist TS-157 through the upregulation of phosphorylated ERK [64].
Stroke-induced damage also compromises blood–brain barrier (BBB) integrity and increases permeability to substances otherwise restricted from the brain [65]. S1R agonism with PRE-084 and dexmedetomidine successfully attenuated this damage by inducing BBB stability through increasing levels of glia-derived neurotrophic factor (GDNF), restoring abnormal expression of the BBB marker Occludin, and decreasing inflammatory markers [58,62,66,67]. Pericyte attachment acts as another contributor to BBB breakdown, which has been shown to improve with S1R activation [68,69]. Post-stroke complications include spreading depolarizations (SDs), which are waves of depolarizations from neural cells due to a failure of ion homeostasis after an insult [70,71]. PRE-084 and DMT have been shown to reduce SD frequency, promoting neural survival and reducing apoptosis [72]. Another feared complication of ischemic stroke includes reperfusion injury, characterized by a paradoxical worsening of damaged cells upon restoration of blood flow, causing inflammation and apoptosis. S1R upregulation appears to attenuate this process and improve outcomes [63,73].
In models of traumatic brain injury (TBI), the role of S1R is complex. In one study, mice deficient in sigma receptors led to better long-term neurological outcomes, such as better coordination and fewer neurological deficits after one-year post-injury [74]. Conversely, other investigations of TBI demonstrate that S1R agonism leads to better neurological function, restoration of blood flow, and decreased brain edema [75]. This apparent biphasic effect may reflect differing impacts of S1R activation depending on timing, with acute-phase activation being beneficial and prolonged activation being potentially detrimental. The acute phase of TBI is marked by inflammation, which is dampened with S1R activation [76]. Although the role of S2R in TBI is less studied, modulation may yield neuroprotective outcomes [77].
In the context of spinal cord injury, S1R activation has demonstrated protective effects in recovery after a mechanical insult to the spine. This was achieved through reducing neuroinflammation, apoptosis, and ferroptosis [78,79]. Ferroptosis is an iron-dependent form of cell death characterized by the buildup of lipid oxidation products. Ferroptosis has been implicated in neuronal cell death after injury and is shown to be upregulated in the setting of spinal cord injury; however, treatment with DMT resulted in marked reductions in this process [72,78,80,81]. The impact of neural injury and the role of S1R is represented in Figure 3.

4. Sigma-1 Receptor in Neurodegenerative Disorders

Sigma receptors have been implicated in a variety of neurodegenerative disorders due to their role in regulating calcium homeostasis, mitochondrial function, and oxidative stress regulation [29,82,83,84]. Genetic polymorphisms in the S1R gene have been associated with increased susceptibility to Alzheimer’s disease (AD), whereas loss-of-function mutations have been linked to the development of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), potentially through mechanisms such as impaired long-term potentiation observed in S1R KO models [85,86,87,88]. Other neurological disorders have had favorable outcomes with sigma-receptor modulation, such as Huntington’s disease (HD), multiple sclerosis (MS), and Parkinson’s disease (PD) [89,90,91]. In recent years, S1R has gained attention as a promising therapeutic target for mitigating these diseases [92]. Additionally, S2R ligands are also emerging as potential modulators of neurodegeneration [93].
S1R agonism confers neuroprotection through several pathways. These include enhanced synaptic plasticity through brain-derived neurotrophic factor (BDNF) dependent mechanisms, reduction of intracellular nitric oxide (NO) by inhibiting NO synthase, and prevention of oxidative stress by reducing the accumulation of reactive oxygen species (ROS) [94,95,96,97,98,99,100,101]. All of these mechanisms support the maintenance of homeostatic plasticity, a process critical to the stabilization of neural pathways, and are central to preventing neurodegeneration [102,103]. Functional evidence of neuroprotection with S1R agonism has been studied in behavioral mouse models using modalities such as novel object recognition, showing improved cognitive outcomes [104,105].
Neuroinflammation also plays a central role in the progression of neurodegenerative disorders [106]. Downregulation of S1R has been associated with increased inflammation markers and disrupted microglial homeostasis [107]. In contrast, S1R agonists and allosteric modulators have been shown to reduce neuroinflammation levels through the reduction of microglial recruitment and inhibition of pro-inflammatory cytokine production, thereby attenuating gliosis and slowing cognitive impairment from various degenerative conditions, such as chronic epilepsy [108,109,110,111].

4.1. Sigma-1 Receptor in Alzheimer’s Disease

Mitochondrial dysfunction is a hallmark of many neurodegenerative disorders, including Alzheimer’s disease (AD) [112]. Restoring mitochondrial stability has become a major therapeutic goal with S1R modulation [113]. In preclinical models, DMT preserved mitochondrial integrity by restoring the expression of MAM-associated proteins. Chronic treatment with DMT not only led to general neuroprotection but also slowed the accumulation of beta-amyloid in the hippocampus, the pathological hallmark of AD [114,115,116,117,118,119]. Similar benefits have been observed with pridopidine and PRE-084, which restored mitochondrial dysfunction by reducing levels of ROS and subsequent astrogliosis as well as stimulating hippocampal proliferation [89,116,118,119,120,121,122,123]. Furthermore, S1R agonism demonstrated synergistic neuroprotective effects when combined with the acetylcholinesterase inhibitor donepezil, the current treatment for AD, again through ROS reduction [100,124].
Cognitive impairment in AD is commonly tested through recognition memory in animal models, which have shown improvement after treatment with the S1R agonists DMT, AF710B, pridopidine, OZP002, LS-1-137, and WLB-87848. AF710B reduced the number of amyloid plaques as well as astrogliosis in the hippocampus. Pridopidine induced the formation of neuronal synapses through activation of the ERK and Akt pathways. OZP002 reduced ROS, apoptotic marker Bax, inflammatory markers TNFα and IL-6, and lipid peroxidation within hippocampal neurons [108,114,117,122,125]. Other S1R agonists, such as dipentylammonium (DPA), did not significantly alter cognitive outcomes but were found to induce neurite growth through increasing levels of NGF and improved lifespan in preclinical models [126,127]. However, some studies have reported paradoxical findings. Specifically, MAM induction has been linked to increased amyloid-beta accumulation, which was reduced with S1R downregulation [128].
Another hallmark of AD pathogenesis is the deposition of neurofibrillary tangles composed of hyperphosphorylated tau protein [129]. In healthy individuals, S1R has been implicated in maintaining proper phosphorylation of tau proteins, and loss of S1R may contribute to tau-related pathology [130,131]. S2R modulation also has a potential role in slowing the progression of AD. CT1812 and SAS-0132, S2R antagonists, displaced amyloid plaques from hippocampal neurons, induced synapses, and regulated tau phosphorylation, improving cognitive performance. WLB-8962, an S2R agonist, improved short-term memory in preclinical models [132,133,134]
Other mechanisms under investigation to slow the progression of AD include the role of preserving BBB integrity. S1R activation has been associated with increasing levels of vascular endothelial growth factor (VEGF) and low-density lipoprotein receptor-related protein 1 (LRP-1), both of which are critical to maintaining BBB function [135]. Newly studied S2R ligands have also demonstrated promising results in beta-amyloid-induced neurological dysfunction, potentially through restoration of calcium homeostasis [133,134]. Figure 4 demonstrates the role of S1R in slowing the progression of AD.
Ongoing clinical trials utilizing S1R or S2R ligands offer promising results for the future of AD treatment. A recent Phase IIb/III trial was conducted in patients with AD to test the efficacy of blarcamesine, an S1R agonist. Blarcamesine significantly slowed cognitive deterioration by 36.3% compared to placebo after 48 weeks. Those with the SIGMAR-1 genotype exhibited the most improvement [136]. Another S1R agonist, T-817MA, was studied in a Phase II trial but did not demonstrate statistically significant reductions in cognitive decline [137]. CT1812 is currently undergoing multiple Phase I and II trials, and a systematic review revealed that Phase I trials demonstrate adequate drug safety and cerebral penetration, opening avenues for future trials to assess their disease-modifying capabilities [138].

4.2. Sigma-1 Receptor in ALS

ALS is a devastating neurodegenerative disease, with the most common inherited form linked to mutations in the C9orf72 gene involving repeats of GGGGCC-RNA [139]. MAM instability and dysregulated autophagy are key pathological features of ALS [140,141,142]. Notably, a common selective serotonin reuptake inhibitor (SSRI), fluvoxamine, possesses S1R agonism and has been shown to restore autophagic balance in inherited ALS by stabilizing nucleoporin complexes with Pom121 protein expression and localizing the autophagy factor TFEB to the nucleus [143].
Additional S1R agonists such as pridopidine, PRE-084, and SA4503 have demonstrated improvements in motor behavior and neuroprotection of ALS mouse models [144,145]. PRE-084, specifically, rescued the mitochondrial dysregulation and ER stress seen in ALS, primarily through increasing the ATF4 and NRF2 antioxidant cascades [121]. Interestingly, in the same experimental settings, BD1063, an S1R antagonist, also yielded neuroprotective effects through a similar mechanism to PRE-084, suggesting a more complex role of S1R signaling in ALS [145].
Another pathological mechanism contributing to the development of ALS involves the dysregulated accumulation of repeat-associated non-AUG (RAN) proteins within neurons. Overexpression of S1R has been shown to reduce RAN protein accumulation, offering a novel therapeutic avenue [146,147]. Additionally, mutations in the Cu/Zn superoxide dismutase (SOD1) gene, another hallmark of familial ALS, can lead to neurofilament accumulation and motor neuron degeneration. Treatment with pridopidine reduced this buildup by activating the ERK pathway, highlighting its potential as a disease-modifying therapy [148,149]. Pridopidine is presently the only sigma receptor ligand undergoing clinical trials for ALS. In a Phase II/III trial studying pridopidine’s ability to alter functional outcomes, respiratory status, and survival in ALS, no significant differences were noted when compared to placebo after 24 weeks [150].

4.3. Sigma-1 Receptor in Huntington’s Disease

HD is an inherited neurodegenerative disorder characterized by progressive neuronal loss, leading to debilitating, uncontrolled movements [151]. As with many neurodegenerative diseases, mitochondrial dysfunction is central to HD pathogenesis [152]. In preclinical models of HD, pridopidine restored the antioxidant defenses and reduced levels of mitochondrial reactive oxygen species through rescuing IP3R localization as well as BDNF, glucocorticoid receptor (GR) dopamine D1 receptor (D1R), cAMP, and TrkB signaling, which restored glutamate homeostasis within the striatum [89,94,153,154]. Pridopidine also restored ER stress through the reduction in the unfolded protein response, which is central to the pathogenesis of HD, most prominently in the PKR-like endoplasmic reticulum kinase (PERK) pathway [155,156]. Additional studies have demonstrated that pridopidine restores calcium homeostasis through the upregulation of calbindin and homer1a, contributing to the attenuation of disease progression [154,156,157,158].
Given that there are currently no approved disease-modifying therapies for HD, the development of novel treatment modalities is urgently needed. There are multiple ongoing clinical trials investigating pridopidine in the treatment of HD. In a review investigating four trials, all in Phase II or III with 1067 total patients with HD, pridopidine was shown to be safe in all cohorts, offering promising treatment modalities for the future [159,160].

4.4. Sigma-1 Receptor in Parkinson’s Disease

PD is one of the most prevalent neurodegenerative disorders, characterized by bradykinesia, tremor, and rigidity [161]. A key pathological feature is the toxic accumulation of alpha-synuclein in the nervous system, which contributes to mitochondrial dysfunction and subsequent neurodegeneration [162]. Deficiencies in S1R exacerbate alpha-synuclein accumulation, highlighting its importance in the pathogenesis of PD [163].
In addition, S2R antagonists have demonstrated neuroprotective effects by mitigating alpha-synuclein-induced toxicity, suggesting a novel therapeutic modality [164]. S1R agonism through PRE-084, either alone or in combination with nicotinic receptor agonists, has shown efficacy in preserving dopaminergic neurons, which are preferentially impacted in PD, as well as providing neuroprotection from MPTP-induced neuronal damage [165,166,167]. Pridopidine was found to improve motor deficits in preclinical models of PD after long-term treatment by increasing the number of dopaminergic neurons in the striatum through upregulation of GDNF, BDNF, and phosphorylated ERK [167]. Levodopa is a mainstay treatment of PD; however, it is frequently associated with dyskinesia as a side effect [168]. Preclinical studies have shown that pridopidine may attenuate levodopa-induced dyskinesia while preserving the benefit [169,170].
S1R antagonists have also demonstrated a protective effect on PD pathogenesis. One study found that S1R downregulates the transient receptor potential canonical (TRPC) channel, which is critical for calcium signaling and maintaining cell viability. S1R antagonism in this context restored TRPC activity and promoted dopaminergic neuroprotection [171]. Additionally, reduced S1R expression has been associated with decreased NMDA-receptor-mediated excitotoxicity, further supporting its complex role in PD [172]. The implications of sigma receptors in modifying the disease state of ALS, HD, and PD are demonstrated in Figure 5.

4.5. Sigma-1 Receptor in Demyelinating Disorders

Demyelinating disorders, both inherited and acquired, have also been investigated in the context of S1R agonism. Krabbe Disease, a rare autosomal recessive leukodystrophy marked by progressive neurodegeneration and demyelination, has shown positive responses to S1R modulation. In animal models, treatment with donepezil, which possesses S1R agonism, preserved myelin integrity and reduced glial reactivity [173,174]. Similar results have been reported in other rare genetic neurodegenerative disorders, such as Wolfram Syndrome and Vanishing White Matter Disease (VWM), where S1R activation demonstrated protection against cellular stress and mitochondrial dysfunction, lowering rates of autophagy [175,176,177,178].
In contrast to these inherited conditions, MS is an acquired autoimmune disease characterized by chronic inflammation, demyelination, and axonal loss [179]. Preclinical studies investigating models of MS concluded that S1R agonists can attenuate the clinical course, offering a promising avenue for clinical translation [110,180]. Specifically, blarcamesine protected oligodendroglia from ROS and quinolinic acid, a common neuroinflammatory marker [90]. The neuroprotective mechanism in MS appears to involve the preservation of oligodendroglia from apoptosis and reactive oxygen species [90]. Presently, there are no current sigma receptor ligands in the clinical stage for demyelinating disorders.

5. Challenges and Future Directions

Despite the significant advances in understanding S1R and its role in neurodegenerative diseases, several challenges limit its clinical translation. Though many mouse models offer promising results in attenuation of neurodegeneration, the precise pathways and downstream targets behind them remain less understood. This significantly limits the ability to evaluate these medications on patients. Additionally, many ligands that have been developed for S1R are rarely selective and may be modulating other pathways that remain undiscovered. The variability in results adds to the difficulty in assessing S1R as a promising target. This adds the need for more context-dependent studies. It will be critical to elucidate the S1R function at various disease stages as well as its tissue-specific expression. The lack of clinical trials, specifically beyond the early phases, leaves S1R undiscovered in the context of its ability to slow neurodegeneration in humans. Despite the progress made, it is imperative to further evaluate the utility of these ligands in a clinical setting.

6. Conclusions

In conclusion, sigma receptors have emerged as key regulators of cellular homeostasis and neuroprotection. Table 1 summarizes key S1R and S2R ligands currently under investigation, including their target diseases, mechanisms, and stage of clinical development. Their broad expression across tissues has driven extensive research into their pharmacologic potential, particularly within the nervous system. Their versatile roles across a variety of pathologies emphasize their relevance as novel therapeutic targets in future research for both acute and chronic neurodegeneration. However, contradictory findings in different models highlight the importance of temporal and tissue-specific modulation to optimize outcomes. Additionally, the integration of S1R into combination therapies may enhance clinical benefit, offering new strategies for previously untreatable diseases.

Author Contributions

Conceptualization, N.D. and D.N.; writing—original draft preparation, N.D. and X.F.; writing—review and editing, D.N. and N.G.; visualization, X.F. and N.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Michael Crider, School of Pharmacy at Southern Illinois University Edwardsville, for critical reading and suggestions of the manuscript. Figures were created with permission from the Servier Medical Art library under the Creative Commons license.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Martin, W.R.; Eades, C.G.; Thompson, J.A.; Huppler, R.E.; Gilbert, P.E. The effects of morphine- and nalorphine- like drugs in the nondependent and morphine-dependent chronic spinal dog. J. Pharmacol. Exp. Ther. 1976, 197, 517–532. [Google Scholar] [CrossRef] [PubMed]
  2. Tam, S.W.; Cook, L. Sigma opiates and certain antipsychotic drugs mutually inhibit (+)-[3H] SKF 10,047 and [3H]haloperidol binding in guinea pig brain membranes. Proc. Natl. Acad. Sci. USA 1984, 81, 5618–5621. [Google Scholar] [CrossRef] [PubMed]
  3. Su, T.P. Evidence for sigma opioid receptor: Binding of [3H]SKF-10047 to etorphine-inaccessible sites in guinea-pig brain. J. Pharmacol. Exp. Ther. 1982, 223, 284–290. [Google Scholar] [CrossRef]
  4. Quirion, R.; Bowen, W.D.; Itzhak, Y.; Junien, J.L.; Musacchio, J.M.; Rothman, R.B.; Su, T.P.; Tam, S.W.; Taylor, D.P. A proposal for the classification of sigma binding sites. Trends Pharmacol. Sci. 1992, 13, 85–86. [Google Scholar] [CrossRef]
  5. Hanner, M.; Moebius, F.F.; Flandorfer, A.; Knaus, H.G.; Striessnig, J.; Kempner, E.; Glossmann, H. Purification, molecular cloning, and expression of the mammalian sigma1-binding site. Proc. Natl. Acad. Sci. USA 1996, 93, 8072–8077. [Google Scholar] [CrossRef]
  6. Alon, A.; Schmidt, H.R.; Wood, M.D.; Sahn, J.J.; Martin, S.F.; Kruse, A.C. Identification of the gene that codes for the sigma(2) receptor. Proc. Natl. Acad. Sci. USA 2017, 114, 7160–7165. [Google Scholar] [CrossRef] [PubMed]
  7. Su, T.P.; Hayashi, T. Understanding the molecular mechanism of sigma-1 receptors: Towards a hypothesis that sigma-1 receptors are intracellular amplifiers for signal transduction. Curr. Med. Chem. 2003, 10, 2073–2080. [Google Scholar] [CrossRef]
  8. Hayashi, T.; Su, T. The sigma receptor: Evolution of the concept in neuropsychopharmacology. Curr. Neuropharmacol. 2005, 3, 267–280. [Google Scholar] [CrossRef]
  9. Palmer, C.P.; Mahen, R.; Schnell, E.; Djamgoz, M.B.; Aydar, E. Sigma-1 receptors bind cholesterol and remodel lipid rafts in breast cancer cell lines. Cancer Res. 2007, 67, 11166–11175. [Google Scholar] [CrossRef]
  10. Prasad, P.D.; Li, H.W.; Fei, Y.J.; Ganapathy, M.E.; Fujita, T.; Plumley, L.H.; Yang-Feng, T.L.; Leibach, F.H.; Ganapathy, V. Exon-intron structure, analysis of promoter region, and chromosomal localization of the human type 1 sigma receptor gene. J. Neurochem. 1998, 70, 443–451. [Google Scholar] [CrossRef]
  11. Kekuda, R.; Prasad, P.D.; Fei, Y.J.; Leibach, F.H.; Ganapathy, V. Cloning and functional expression of the human type 1 sigma receptor (hSigmaR1). Biochem. Biophys. Res. Commun. 1996, 229, 553–558. [Google Scholar] [CrossRef] [PubMed]
  12. Seth, P.; Leibach, F.H.; Ganapathy, V. Cloning and structural analysis of the cDNA and the gene encoding the murine type 1 sigma receptor. Biochem. Biophys. Res. Commun. 1997, 241, 535–540. [Google Scholar] [CrossRef] [PubMed]
  13. Schmidt, H.R.; Zheng, S.; Gurpinar, E.; Koehl, A.; Manglik, A.; Kruse, A.C. Crystal structure of the human σ1 receptor. Nature 2016, 532, 527–530. [Google Scholar] [CrossRef]
  14. Waterhouse, A.; Bertoni, M.; Bienert, S.; Studer, G.; Tauriello, G.; Gumienny, R.; Heer, F.T.; de Beer, T.A.P.; Rempfer, C.; Bordoli, L.; et al. SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res. 2018, 46, W296–W303. [Google Scholar] [CrossRef] [PubMed]
  15. Bertoni, M.; Kiefer, F.; Biasini, M.; Bordoli, L.; Schwede, T. Modeling protein quaternary structure of homo- and hetero-oligomers beyond binary interactions by homology. Sci. Rep. 2017, 7, 10480. [Google Scholar] [CrossRef]
  16. Hong, W.C. Distinct Regulation of sigma (1) Receptor Multimerization by Its Agonists and Antagonists in Transfected Cells and Rat Liver Membranes. J. Pharmacol. Exp. Ther. 2020, 373, 290–301. [Google Scholar] [CrossRef]
  17. Navarro, G.; Moreno, E.; Bonaventura, J.; Brugarolas, M.; Farré, D.; Aguinaga, D.; Mallol, J.; Cortés, A.; Casadó, V.; Lluís, C.; et al. Cocaine inhibits dopamine D2 receptor signaling via sigma-1-D2 receptor heteromers. PLoS ONE 2013, 8, e61245. [Google Scholar] [CrossRef]
  18. Aguinaga, D.; Medrano, M.; Vega-Quiroga, I.; Gysling, K.; Canela, E.I.; Navarro, G.; Franco, R. Cocaine Effects on Dopaminergic Transmission Depend on a Balance between Sigma-1 and Sigma-2 Receptor Expression. Front. Mol. Neurosci. 2018, 11, 17. [Google Scholar] [CrossRef]
  19. Fontanilla, D.; Hajipour, A.R.; Pal, A.; Chu, U.B.; Arbabian, M.; Ruoho, A.E. Probing the steroid binding domain-like I (SBDLI) of the sigma-1 receptor binding site using N-substituted photoaffinity labels. Biochemistry 2008, 47, 7205–7217. [Google Scholar] [CrossRef]
  20. Hayashi, T.; Su, T.P. 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, 718–725. [Google Scholar] [CrossRef]
  21. Hayashi, T.; Su, T.P. Sigma-1 receptors at galactosylceramide-enriched lipid microdomains regulate oligodendrocyte differentiation. Proc. Natl. Acad. Sci. USA 2004, 101, 14949–14954. [Google Scholar] [CrossRef] [PubMed]
  22. Gebreselassie, D.; Bowen, W.D. Sigma-2 receptors are specifically localized to lipid rafts in rat liver membranes. Eur. J. Pharmacol. 2004, 493, 19–28. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, J.; Zeng, C.; Chu, W.; Pan, F.; Rothfuss, J.M.; Zhang, F.; Tu, Z.; Zhou, D.; Zeng, D.; Vangveravong, S.; et al. Identification of the PGRMC1 protein complex as the putative sigma-2 receptor binding site. Nat. Commun. 2011, 2, 380. [Google Scholar] [CrossRef] [PubMed]
  24. Vaupel, D.B. Naltrexone fails to antagonize the sigma effects of PCP and SKF 10,047 in the dog. Eur. J. Pharmacol. 1983, 92, 269–274. [Google Scholar] [CrossRef]
  25. Chu, U.B.; Ruoho, A.E. Sigma Receptor Binding Assays. Curr. Protoc. Pharmacol. 2015, 71, 1–34. [Google Scholar] [CrossRef]
  26. Hayashi, T.; Su, T.P. Intracellular dynamics of sigma-1 receptors (sigma(1) binding sites) in NG108-15 cells. J. Pharmacol. Exp. Ther. 2003, 306, 726–733. [Google Scholar] [CrossRef]
  27. Hayashi, T.; Su, T.P. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell 2007, 131, 596–610. [Google Scholar] [CrossRef]
  28. Aydar, E.; Palmer, C.P.; Klyachko, V.A.; Jackson, M.B. The sigma receptor as a ligand-regulated auxiliary potassium channel subunit. Neuron 2002, 34, 399–410. [Google Scholar] [CrossRef]
  29. Su, T.P.; Hayashi, T.; Maurice, T.; Buch, S.; Ruoho, A.E. The sigma-1 receptor chaperone as an inter-organelle signaling modulator. Trends Pharmacol. Sci. 2010, 31, 557–566. [Google Scholar] [CrossRef]
  30. Brimson, J.M.; Brown, C.A.; Safrany, S.T. Antagonists show GTP-sensitive high-affinity binding to the sigma-1 receptor. Br. J. Pharmacol. 2011, 164, 772–780. [Google Scholar] [CrossRef]
  31. Sánchez-Blázquez, P.; Rodríguez-Muñoz, M.; Herrero-Labrador, R.; Burgueño, J.; Zamanillo, D.; Garzón, J. 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, 1943–1955. [Google Scholar] [CrossRef] [PubMed]
  32. Vilner, B.J.; John, C.S.; Bowen, W.D. Sigma-1 and sigma-2 receptors are expressed in a wide variety of human and rodent tumor cell lines. Cancer Res. 1995, 55, 408–413. [Google Scholar] [PubMed]
  33. Bermack, J.E.; Debonnel, G. Distinct modulatory roles of sigma receptor subtypes on glutamatergic responses in the dorsal hippocampus. Synapse 2005, 55, 37–44. [Google Scholar] [CrossRef] [PubMed]
  34. Cobos, E.J.; Entrena, J.M.; Nieto, F.R.; Cendán, C.M.; Del Pozo, E. Pharmacology and therapeutic potential of sigma(1) receptor ligands. Curr. Neuropharmacol. 2008, 6, 344–366. [Google Scholar] [CrossRef]
  35. Hayashi, T.; Maurice, T.; Su, T.P. Ca(2+) signaling via sigma(1)-receptors: Novel regulatory mechanism affecting intracellular Ca(2+) concentration. J. Pharmacol. Exp. Ther. 2000, 293, 788–798. [Google Scholar] [CrossRef]
  36. Hayashi, T.; Su, T.P. Regulating ankyrin dynamics: Roles of sigma-1 receptors. Proc. Natl. Acad. Sci. USA 2001, 98, 491–496. [Google Scholar] [CrossRef]
  37. Hayashi, T.; Su, T.P. Sigma-1 receptor ligands: Potential in the treatment of neuropsychiatric disorders. CNS Drugs 2004, 18, 269–284. [Google Scholar] [CrossRef]
  38. Hayashi, T.; Fujimoto, M. Detergent-resistant microdomains determine the localization of sigma-1 receptors to the endoplasmic reticulum-mitochondria junction. Mol. Pharmacol. 2010, 77, 517–528. [Google Scholar] [CrossRef]
  39. Carnally, S.M.; Johannessen, M.; Henderson, R.M.; Jackson, M.B.; Edwardson, J.M. Demonstration of a direct interaction between sigma-1 receptors and acid-sensing ion channels. Biophys. J. 2010, 98, 1182–1191. [Google Scholar] [CrossRef]
  40. Crottès, D.; Martial, S.; Rapetti-Mauss, R.; Pisani, D.F.; Loriol, C.; Pellissier, B.; Martin, P.; Chevet, E.; Borgese, F.; Soriani, O. Sig1R protein regulates hERG channel expression through a post-translational mechanism in leukemic cells. J. Biol. Chem. 2011, 286, 27947–27958. [Google Scholar] [CrossRef]
  41. Balasuriya, D.; Stewart, A.P.; Crottès, D.; Borgese, F.; Soriani, O.; Edwardson, J.M. The sigma-1 receptor binds to the Nav1.5 voltage-gated Na+ channel with 4-fold symmetry. J. Biol. Chem. 2012, 287, 37021–37029. [Google Scholar] [CrossRef] [PubMed]
  42. Kourrich, S.; Hayashi, T.; Chuang, J.Y.; Tsai, S.Y.; Su, T.P.; Bonci, A. Dynamic interaction between sigma-1 receptor and Kv1.2 shapes neuronal and behavioral responses to cocaine. Cell 2013, 152, 236–247. [Google Scholar] [CrossRef] [PubMed]
  43. Burnett, M.G.; Zager, E.L. Pathophysiology of peripheral nerve injury: A brief review. Neurosurg. Focus 2004, 16, E1. [Google Scholar] [CrossRef]
  44. Lee, R.H.C.; Lee, M.H.H.; Wu, C.Y.C.; Couto, E.S.A.; Possoit, H.E.; Hsieh, T.H.; Minagar, A.; Lin, H.W. Cerebral ischemia and neuroregeneration. Neural Regen. Res. 2018, 13, 373–385. [Google Scholar] [CrossRef]
  45. Ng, S.Y.; Lee, A.Y.W. Traumatic Brain Injuries: Pathophysiology and Potential Therapeutic Targets. Front. Cell. Neurosci. 2019, 13, 528. [Google Scholar] [CrossRef] [PubMed]
  46. Schonfeld, E.; Johnstone, T.M.; Haider, G.; Shah, A.; Marianayagam, N.J.; Biswal, S.; Veeravagu, A. Sigma-1 receptor expression in a subpopulation of lumbar spinal cord microglia in response to peripheral nerve injury. Sci. Rep. 2023, 13, 14762. [Google Scholar] [CrossRef]
  47. Nardai, S.; László, M.; Szabó, A.; Alpár, A.; Hanics, J.; Zahola, P.; Merkely, B.; Frecska, E.; Nagy, Z. N,N-dimethyltryptamine reduces infarct size and improves functional recovery following transient focal brain ischemia in rats. Exp. Neurol. 2020, 327, 113245. [Google Scholar] [CrossRef]
  48. Zhai, M.; Liu, C.; Li, Y.; Zhang, P.; Yu, Z.; Zhu, H.; Zhang, L.; Zhang, Q.; Wang, J.; Wang, J. Dexmedetomidine inhibits neuronal apoptosis by inducing Sigma-1 receptor signaling in cerebral ischemia-reperfusion injury. Aging 2019, 11, 9556–9568. [Google Scholar] [CrossRef]
  49. Morihara, R.; Yamashita, T.; Liu, X.; Nakano, Y.; Fukui, Y.; Sato, K.; Ohta, Y.; Hishikawa, N.; Shang, J.; Abe, K. Protective effect of a novel sigma-1 receptor agonist is associated with reduced endoplasmic reticulum stress in stroke male mice. J. Neurosci. Res. 2018, 96, 1707–1716. [Google Scholar] [CrossRef]
  50. Lozano, R.; Naghavi, M.; Foreman, K.; Lim, S.; Shibuya, K.; Aboyans, V.; Abraham, J.; Adair, T.; Aggarwal, R.; Ahn, S.Y.; et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2095–2128. [Google Scholar] [CrossRef]
  51. Saunders, D.E.; Clifton, A.G.; Brown, M.M. Measurement of infarct size using MRI predicts prognosis in middle cerebral artery infarction. Stroke 1995, 26, 2272–2276. [Google Scholar] [CrossRef] [PubMed]
  52. Schreihofer, D.A.; Dalwadi, D.; Kim, S.; Metzger, D.; Oppong-Gyebi, A.; Das-Earl, P.; Schetz, J.A. Treatment of Stroke at a Delayed Timepoint with a Repurposed Drug Targeting Sigma 1 Receptors. Transl. Stroke Res. 2024, 15, 1035–1049. [Google Scholar] [CrossRef]
  53. van der Heijden, K.V.; Zuiker, R.; Otto, M.E.; Bryan, C.S.; Stewart, N.; Stillwell, C.; De Kam, M.L.; van Leuken, M.B.; van Gerven, J.M.A.; Jacobs, G.E. Safety, Pharmacokinetics, and Pharmacodynamics of a 6-h N,N-Dimethyltryptamine (DMT) Infusion in Healthy Volunteers: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Transl. Sci. 2025, 18, e70234. [Google Scholar] [CrossRef] [PubMed]
  54. Gao, Y.; Liang, W.; Hu, X.; Zhang, W.; Stetler, R.A.; Vosler, P.; Cao, G.; Chen, J. Neuroprotection against hypoxic-ischemic brain injury by inhibiting the apoptotic protease activating factor-1 pathway. Stroke 2010, 41, 166–172. [Google Scholar] [CrossRef]
  55. Katnik, C.; Garcia, A.; Behensky, A.A.; Yasny, I.E.; Shuster, A.M.; Seredenin, S.B.; Petrov, A.V.; Cuevas, J. Activation of σ1 and σ2 receptors by afobazole increases glial cell survival and prevents glial cell activation and nitrosative stress after ischemic stroke. J. Neurochem. 2016, 139, 497–509. [Google Scholar] [CrossRef]
  56. Huang, L.; Wu, Z.B.; Zhuge, Q.; Zheng, W.; Shao, B.; Wang, B.; Sun, F.; Jin, K. Glial scar formation occurs in the human brain after ischemic stroke. Int. J. Med. Sci. 2014, 11, 344–348. [Google Scholar] [CrossRef]
  57. Sánchez-Blázquez, P.; Pozo-Rodrigálvarez, A.; Merlos, M.; Garzón, J. The Sigma-1 Receptor Antagonist, S1RA, Reduces Stroke Damage, Ameliorates Post-Stroke Neurological Deficits and Suppresses the Overexpression of MMP-9. Mol. Neurobiol. 2018, 55, 4940–4951. [Google Scholar] [CrossRef]
  58. Song, W.; Yao, Y.; Zhang, H.; Hao, X.; Zhou, L.; Song, Z.; Wei, T.; Chi, T.; Liu, P.; Ji, X.; et al. Sigma-1 Receptor Activation Improves Oligodendrogenesis and Promotes White-Matter Integrity after Stroke in Mice with Diabetic Mellitus. Molecules 2023, 28, 390. [Google Scholar] [CrossRef] [PubMed]
  59. Zhang, W.; Zhao, J.; Wang, R.; Jiang, M.; Ye, Q.; Smith, A.D.; Chen, J.; Shi, Y. Macrophages reprogram after ischemic stroke and promote efferocytosis and inflammation resolution in the mouse brain. CNS Neurosci. Ther. 2019, 25, 1329–1342. [Google Scholar] [CrossRef]
  60. Zhang, G.; Li, Q.; Tao, W.; Qin, P.; Chen, J.; Yang, H.; Chen, J.; Liu, H.; Dai, Q.; Zhen, X. Sigma-1 receptor-regulated efferocytosis by infiltrating circulating macrophages/microglial cells protects against neuronal impairments and promotes functional recovery in cerebral ischemic stroke. Theranostics 2023, 13, 543–559. [Google Scholar] [CrossRef]
  61. Yabuki, Y.; Shinoda, Y.; Izumi, H.; Ikuno, T.; Shioda, N.; Fukunaga, K. Dehydroepiandrosterone administration improves memory deficits following transient brain ischemia through sigma-1 receptor stimulation. Brain Res. 2015, 1622, 102–113. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, D.Y.; Chi, T.Y.; Ji, X.F.; Liu, P.; Qi, X.X.; Zhu, L.; Wang, Z.Q.; Li, L.; Chen, L.; Zou, L.B. Sigma-1 receptor activation alleviates blood-brain barrier dysfunction in vascular dementia mice. Exp. Neurol. 2018, 308, 90–99. [Google Scholar] [CrossRef]
  63. Dai, L.A.; Chen, X.Y.; Li, W.J.; Yang, J.H.; Lin, M.J.; Li, X.S.; Zeng, Y.F.; Chen, S.W.; Xie, Z.L.; Zhu, Z.L.; et al. Sigma-1 Receptor and Binding Immunoglobulin Protein Interact with Ulinastatin Contributing to a Protective Effect in Rat Cerebral Ischemia/Reperfusion. World Neurosurg. 2022, 158, e488–e494. [Google Scholar] [CrossRef] [PubMed]
  64. Shi, J.J.; Jiang, Q.H.; Zhang, T.N.; Sun, H.; Shi, W.W.; Gunosewoyo, H.; Yang, F.; Tang, J.; Pang, T.; Yu, L.F. Sigma-1 Receptor Agonist TS-157 Improves Motor Functional Recovery by Promoting Neurite Outgrowth and pERK in Rats with Focal Cerebral Ischemia. Molecules 2021, 26, 1212. [Google Scholar] [CrossRef]
  65. Okada, T.; Suzuki, H.; Travis, Z.D.; Zhang, J.H. The Stroke-Induced Blood-Brain Barrier Disruption: Current Progress of Inspection Technique, Mechanism, and Therapeutic Target. Curr. Neuropharmacol. 2020, 18, 1187–1212. [Google Scholar] [CrossRef]
  66. Liu, D.; Yang, L.; Liu, P.; Ji, X.; Qi, X.; Wang, Z.; Chi, T.; Zou, L. Sigma-1 receptor activation alleviates blood-brain barrier disruption post cerebral ischemia stroke by stimulating the GDNF-GFRα1-RET pathway. Exp. Neurol. 2022, 347, 113867. [Google Scholar] [CrossRef] [PubMed]
  67. Zhao, Q.; Yu, S.; Ling, Y.; Hao, S.; Liu, J. The Protective Effects of Dexmedetomidine against Hypoxia/Reoxygenation-Induced Inflammatory Injury and Permeability in Brain Endothelial Cells Mediated by Sigma-1 Receptor. ACS Chem. Neurosci. 2021, 12, 1940–1947. [Google Scholar] [CrossRef]
  68. Yang, S.; Jin, H.; Zhu, Y.; Wan, Y.; Opoku, E.N.; Zhu, L.; Hu, B. Diverse Functions and Mechanisms of Pericytes in Ischemic Stroke. Curr. Neuropharmacol. 2017, 15, 892–905. [Google Scholar] [CrossRef]
  69. Zhang, Y.; Zhang, X.; Wei, Q.; Leng, S.; Li, C.; Han, B.; Bai, Y.; Zhang, H.; Yao, H. Activation of Sigma-1 Receptor Enhanced Pericyte Survival via the Interplay Between Apoptosis and Autophagy: Implications for Blood-Brain Barrier Integrity in Stroke. Transl. Stroke Res. 2020, 11, 267–287. [Google Scholar] [CrossRef]
  70. Dohmen, C.; Sakowitz, O.W.; Fabricius, M.; Bosche, B.; Reithmeier, T.; Ernestus, R.I.; Brinker, G.; Dreier, J.P.; Woitzik, J.; Strong, A.J.; et al. Spreading depolarizations occur in human ischemic stroke with high incidence. Ann. Neurol. 2008, 63, 720–728. [Google Scholar] [CrossRef]
  71. Leao, A.A. The slow voltage variation of cortical spreading depression of activity. Electroencephalogr. Clin. Neurophysiol. 1951, 3, 315–321. [Google Scholar] [CrossRef] [PubMed]
  72. Szabó, Í.; Varga, V.; Dvorácskó, S.; Farkas, A.E.; Körmöczi, T.; Berkecz, R.; Kecskés, S.; Menyhárt, Á.; Frank, R.; Hantosi, D.; et al. N,N-Dimethyltryptamine attenuates spreading depolarization and restrains neurodegeneration by sigma-1 receptor activation in the ischemic rat brain. Neuropharmacology 2021, 192, 108612. [Google Scholar] [CrossRef]
  73. Soares, R.O.S.; Losada, D.M.; Jordani, M.C.; Evora, P.; Castro, E.S.O. Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies. Int. J. Mol. Sci. 2019, 20, 5034. [Google Scholar] [CrossRef] [PubMed]
  74. Stelfa, G.; Vavers, E.; Svalbe, B.; Serzants, R.; Miteniece, A.; Lauberte, L.; Grinberga, S.; Gukalova, B.; Dambrova, M.; Zvejniece, L. Reduced GFAP Expression in Bergmann Glial Cells in the Cerebellum of Sigma-1 Receptor Knockout Mice Determines the Neurobehavioral Outcomes after Traumatic Brain Injury. Int. J. Mol. Sci. 2021, 22, 11611. [Google Scholar] [CrossRef]
  75. Shi, M.; Liu, L.; Min, X.; Mi, L.; Chai, Y.; Chen, F.; Wang, J.; Yue, S.; Zhang, J.; Deng, Q.; et al. Activation of Sigma-1 Receptor Alleviates ER-Associated Cell Death and Microglia Activation in Traumatically Injured Mice. J. Clin. Med. 2022, 11, 2348. [Google Scholar] [CrossRef]
  76. Dong, H.; Ma, Y.; Ren, Z.; Xu, B.; Zhang, Y.; Chen, J.; Yang, B. Sigma-1 Receptor Modulates Neuroinflammation After Traumatic Brain Injury. Cell. Mol. Neurobiol. 2016, 36, 639–645. [Google Scholar] [CrossRef]
  77. Vázquez-Rosa, E.; Watson, M.R.; Sahn, J.J.; Hodges, T.R.; Schroeder, R.E.; Cintrón-Pérez, C.J.; Shin, M.K.; Yin, T.C.; Emery, J.L.; Martin, S.F.; et al. Neuroprotective Efficacy of a Sigma 2 Receptor/TMEM97 Modulator (DKR-1677) after Traumatic Brain Injury. ACS Chem. Neurosci. 2019, 10, 1595–1602. [Google Scholar] [CrossRef] [PubMed]
  78. Tan, R.; Sui, C.; Diao, Y.; Shi, G.; Hu, X.; Hao, Z.; Li, C.; Hao, M.; Xie, M.; Zhu, T. Activation of the sigma-1 receptor ameliorates neuronal ferroptosis via IRE1α after spinal cord injury. Brain Res. 2024, 1838, 149011. [Google Scholar] [CrossRef]
  79. Gaja-Capdevila, N.; Hernández, N.; Yeste, S.; Reinoso, R.F.; Burgueño, J.; Montero, A.; Merlos, M.; Vela, J.M.; Herrando-Grabulosa, M.; Navarro, X. EST79232 and EST79376, Two Novel Sigma-1 Receptor Ligands, Exert Neuroprotection on Models of Motoneuron Degeneration. Int. J. Mol. Sci. 2022, 23, 6737. [Google Scholar] [CrossRef]
  80. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  81. Kenny, E.M.; Fidan, E.; Yang, Q.; Anthonymuthu, T.S.; New, L.A.; Meyer, E.A.; Wang, H.; Kochanek, P.M.; Dixon, C.E.; Kagan, V.E.; et al. Ferroptosis Contributes to Neuronal Death and Functional Outcome After Traumatic Brain Injury. Crit. Care Med. 2019, 47, 410–418. [Google Scholar] [CrossRef] [PubMed]
  82. Maurice, T.; Su, T.P. The pharmacology of sigma-1 receptors. Pharmacol. Ther. 2009, 124, 195–206. [Google Scholar] [CrossRef] [PubMed]
  83. Urani, A.; Romieu, P.; Roman, F.J.; Maurice, T. Enhanced antidepressant effect of sigma(1) (sigma(1)) receptor agonists in beta(25-35)-amyloid peptide-treated mice. Behav. Brain Res. 2002, 134, 239–247. [Google Scholar] [CrossRef]
  84. Genovese, I.; Giamogante, F.; Barazzuol, L.; Battista, T.; Fiorillo, A.; Vicario, M.; D’Alessandro, G.; Cipriani, R.; Limatola, C.; Rossi, D.; et al. Sorcin is an early marker of neurodegeneration, Ca(2+) dysregulation and endoplasmic reticulum stress associated to neurodegenerative diseases. Cell Death Dis. 2020, 11, 861. [Google Scholar] [CrossRef]
  85. Luty, A.A.; Kwok, J.B.; Dobson-Stone, C.; Loy, C.T.; Coupland, K.G.; Karlström, H.; Sobow, T.; Tchorzewska, J.; Maruszak, A.; Barcikowska, M.; et al. Sigma nonopioid intracellular receptor 1 mutations cause frontotemporal lobar degeneration-motor neuron disease. Ann. Neurol. 2010, 68, 639–649. [Google Scholar] [CrossRef]
  86. Al-Saif, A.; Al-Mohanna, F.; Bohlega, S. A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann. Neurol. 2011, 70, 913–919. [Google Scholar] [CrossRef] [PubMed]
  87. Huang, Y.; Zheng, L.; Halliday, G.; Dobson-Stone, C.; Wang, Y.; Tang, H.D.; Cao, L.; Deng, Y.L.; Wang, G.; Zhang, Y.M.; et al. Genetic polymorphisms in sigma-1 receptor and apolipoprotein E interact to influence the severity of Alzheimer’s disease. Curr. Alzheimer Res. 2011, 8, 765–770. [Google Scholar] [CrossRef]
  88. Snyder, M.A.; McCann, K.; Lalande, M.J.; Thivierge, J.P.; Bergeron, R. Sigma receptor type 1 knockout mice show a mild deficit in plasticity but no significant change in synaptic transmission in the CA1 region of the hippocampus. J. Neurochem. 2016, 138, 700–709. [Google Scholar] [CrossRef]
  89. Naia, L.; Ly, P.; Mota, S.I.; Lopes, C.; Maranga, C.; Coelho, P.; Gershoni-Emek, N.; Ankarcrona, M.; Geva, M.; Hayden, M.R.; et al. The Sigma-1 Receptor Mediates Pridopidine Rescue of Mitochondrial Function in Huntington Disease Models. Neurotherapeutics 2021, 18, 1017–1038. [Google Scholar] [CrossRef]
  90. Lisak, R.P.; Nedelkoska, L.; Benjamins, J.A. Sigma-1 receptor agonists as potential protective therapies in multiple sclerosis. J. Neuroimmunol. 2020, 342, 577188. [Google Scholar] [CrossRef]
  91. Francardo, V. Sigma-1 receptor: A potential new target for Parkinson’s disease? Neural Regen. Res. 2014, 9, 1882–1883. [Google Scholar] [CrossRef] [PubMed]
  92. Nguyen, L.; Kaushal, N.; Robson, M.J.; Matsumoto, R.R. Sigma receptors as potential therapeutic targets for neuroprotection. Eur. J. Pharmacol. 2014, 743, 42–47. [Google Scholar] [CrossRef] [PubMed]
  93. Mondal, S.; Hegarty, E.; Sahn, J.J.; Scott, L.L.; Gökçe, S.K.; Martin, C.; Ghorashian, N.; Satarasinghe, P.N.; Iyer, S.; Sae-Lee, W.; et al. High-Content Microfluidic Screening Platform Used To Identify σ2R/Tmem97 Binding Ligands that Reduce Age-Dependent Neurodegeneration in C. elegans SC_APP Model. ACS Chem. Neurosci. 2018, 9, 1014–1026. [Google Scholar] [CrossRef] [PubMed]
  94. Lenoir, S.; Lahaye, R.A.; Vitet, H.; Scaramuzzino, C.; Virlogeux, A.; Capellano, L.; Genoux, A.; Gershoni-Emek, N.; Geva, M.; Hayden, M.R.; et al. Pridopidine rescues BDNF/TrkB trafficking dynamics and synapse homeostasis in a Huntington disease brain-on-a-chip model. Neurobiol. Dis. 2022, 173, 105857. [Google Scholar] [CrossRef]
  95. Geva, M.; Kusko, R.; Soares, H.; Fowler, K.D.; Birnberg, T.; Barash, S.; Wagner, A.M.; Fine, T.; Lysaght, A.; Weiner, B.; et al. Pridopidine activates neuroprotective pathways impaired in Huntington Disease. Hum. Mol. Genet. 2016, 25, 3975–3987. [Google Scholar] [CrossRef]
  96. Lu, B.; Nagappan, G.; Guan, X.; Nathan, P.J.; Wren, P. BDNF-based synaptic repair as a disease-modifying strategy for neurodegenerative diseases. Nat. Rev. Neurosci. 2013, 14, 401–416. [Google Scholar] [CrossRef]
  97. Goyagi, T.; Goto, S.; Bhardwaj, A.; Dawson, V.L.; Hurn, P.D.; Kirsch, J.R. Neuroprotective effect of sigma(1)-receptor ligand 4-phenyl-1-(4-phenylbutyl) piperidine (PPBP) is linked to reduced neuronal nitric oxide production. Stroke 2001, 32, 1613–1620. [Google Scholar] [CrossRef]
  98. Vagnerova, K.; Hurn, P.D.; Bhardwaj, A.; Kirsch, J.R. Sigma 1 receptor agonists act as neuroprotective drugs through inhibition of inducible nitric oxide synthase. Anesth. Analg. 2006, 103, 430–434, table of contents. [Google Scholar] [CrossRef]
  99. Zampieri, D.; Fortuna, S.; Calabretti, A.; Romano, M.; Menegazzi, R.; Schepmann, D.; Wünsch, B.; Collina, S.; Zanon, D.; Mamolo, M.G. Discovery of new potent dual sigma receptor/GluN2b ligands with antioxidant property as neuroprotective agents. Eur. J. Med. Chem. 2019, 180, 268–282. [Google Scholar] [CrossRef]
  100. Rui, M.; Rossino, G.; Coniglio, S.; Monteleone, S.; Scuteri, A.; Malacrida, A.; Rossi, D.; Catenacci, L.; Sorrenti, M.; Paolillo, M.; et al. Identification of dual Sigma1 receptor modulators/acetylcholinesterase inhibitors with antioxidant and neurotrophic properties, as neuroprotective agents. Eur. J. Med. Chem. 2018, 158, 353–370. [Google Scholar] [CrossRef]
  101. Su, T.C.; Lin, S.H.; Lee, P.T.; Yeh, S.H.; Hsieh, T.H.; Chou, S.Y.; Su, T.P.; Hung, J.J.; Chang, W.C.; Lee, Y.C.; et al. The sigma-1 receptor-zinc finger protein 179 pathway protects against hydrogen peroxide-induced cell injury. Neuropharmacology 2016, 105, 1–9. [Google Scholar] [CrossRef] [PubMed]
  102. Turrigiano, G. Homeostatic synaptic plasticity: Local and global mechanisms for stabilizing neuronal function. Cold Spring Harb. Perspect. Biol. 2012, 4, a005736. [Google Scholar] [CrossRef]
  103. Pozo, K.; Goda, Y. Unraveling mechanisms of homeostatic synaptic plasticity. Neuron 2010, 66, 337–351. [Google Scholar] [CrossRef] [PubMed]
  104. Szczepańska, K.; Bojarski, A.J.; Popik, P.; Malikowska-Racia, N. Novel object recognition test as an alternative approach to assessing the pharmacological profile of sigma-1 receptor ligands. Pharmacol. Rep. 2023, 75, 1291–1298. [Google Scholar] [CrossRef] [PubMed]
  105. Sahlholm, K.; Valle-León, M.; Fernández-Dueñas, V.; Ciruela, F. Pridopidine Reverses Phencyclidine-Induced Memory Impairment. Front. Pharmacol. 2018, 9, 338. [Google Scholar] [CrossRef]
  106. Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  107. Iwamoto, M.; Nakamura, Y.; Takemura, M.; Hisaoka-Nakashima, K.; Morioka, N. TLR4-TAK1-p38 MAPK pathway and HDAC6 regulate the expression of sigma-1 receptors in rat primary cultured microglia. J. Pharmacol. Sci. 2020, 144, 23–29. [Google Scholar] [CrossRef]
  108. Orciani, C.; Do Carmo, S.; Foret, M.K.; Hall, H.; Bonomo, Q.; Lavagna, A.; Huang, C.; Cuello, A.C. Early treatment with an M1 and sigma-1 receptor agonist prevents cognitive decline in a transgenic rat model displaying Alzheimer-like amyloid pathology. Neurobiol. Aging 2023, 132, 220–232. [Google Scholar] [CrossRef]
  109. Ji, J.; Gao, C.; Wang, Q.; Jia, X.; Tian, H.; Wei, Y.; Liu, Z.; Wang, Y.; Guo, L. The sigma-1 receptor-TAMM41 axis modulates neuroinflammation and attenuates memory impairment during the latent period of epileptogenesis. Anim. Model Exp. Med. 2025, 8, 197–208. [Google Scholar] [CrossRef]
  110. Guo, L.; Gao, T.; Jia, X.; Gao, C.; Tian, H.; Wei, Y.; Lu, W.; Liu, Z.; Wang, Y. SKF83959 Attenuates Memory Impairment and Depressive-like Behavior during the Latent Period of Epilepsy via Allosteric Activation of the Sigma-1 Receptor. ACS Chem. Neurosci. 2022, 13, 3198–3209. [Google Scholar] [CrossRef]
  111. Heiss, K.; Raffaele, M.; Vanella, L.; Murabito, P.; Prezzavento, O.; Marrazzo, A.; Aricò, G.; Castracani, C.C.; Barbagallo, I.; Zappalà, A.; et al. (+)-Pentazocine attenuates SH-SY5Y cell death, oxidative stress and microglial migration induced by conditioned medium from activated microglia. Neurosci. Lett. 2017, 642, 86–90. [Google Scholar] [CrossRef] [PubMed]
  112. Johri, A.; Beal, M.F. Mitochondrial dysfunction in neurodegenerative diseases. J. Pharmacol. Exp. Ther. 2012, 342, 619–630. [Google Scholar] [CrossRef] [PubMed]
  113. Cavendish, J.Z.; Sarkar, S.N.; Colantonio, M.A.; Quintana, D.D.; Ahmed, N.; White, B.A.; Engler-Chiurazzi, E.B.; Simpkins, J.W. Mitochondrial Movement and Number Deficits in Embryonic Cortical Neurons from 3xTg-AD Mice. J. Alzheimer’s Dis. 2019, 70, 139–151. [Google Scholar] [CrossRef] [PubMed]
  114. Cheng, D.; Lei, Z.G.; Chu, K.; Lam, O.J.H.; Chiang, C.Y.; Zhang, Z.J. N, N-Dimethyltryptamine, a natural hallucinogen, ameliorates Alzheimer’s disease by restoring neuronal Sigma-1 receptor-mediated endoplasmic reticulum-mitochondria crosstalk. Alzheimer’s Res. Ther. 2024, 16, 95. [Google Scholar] [CrossRef]
  115. Murphy, M.P.; LeVine, H., 3rd. Alzheimer’s disease and the amyloid-beta peptide. J. Alzheimer’s Dis. 2010, 19, 311–323. [Google Scholar] [CrossRef]
  116. Borbély, E.; Varga, V.; Szögi, T.; Schuster, I.; Bozsó, Z.; Penke, B.; Fülöp, L. Impact of Two Neuronal Sigma-1 Receptor Modulators, PRE084 and DMT, on Neurogenesis and Neuroinflammation in an Aβ(1-42)-Injected, Wild-Type Mouse Model of AD. Int. J. Mol. Sci. 2022, 23, 2514. [Google Scholar] [CrossRef]
  117. Maurice, T.; Volle, J.N.; Strehaiano, M.; Crouzier, L.; Pereira, C.; Kaloyanov, N.; Virieux, D.; Pirat, J.L. Neuroprotection in non-transgenic and transgenic mouse models of Alzheimer’s disease by positive modulation of σ(1) receptors. Pharmacol. Res. 2019, 144, 315–330. [Google Scholar] [CrossRef]
  118. Hall, H.; Iulita, M.F.; Gubert, P.; Flores Aguilar, L.; Ducatenzeiler, A.; Fisher, A.; Cuello, A.C. AF710B, an M1/sigma-1 receptor agonist with long-lasting disease-modifying properties in a transgenic rat model of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 811–823. [Google Scholar] [CrossRef]
  119. Fisher, A.; Bezprozvanny, I.; Wu, L.; Ryskamp, D.A.; Bar-Ner, N.; Natan, N.; Brandeis, R.; Elkon, H.; Nahum, V.; Gershonov, E.; et al. AF710B, a Novel M1/σ1 Agonist with Therapeutic Efficacy in Animal Models of Alzheimer’s Disease. Neurodegener. Dis. 2016, 16, 95–110. [Google Scholar] [CrossRef]
  120. Uttara, B.; Singh, A.V.; Zamboni, P.; Mahajan, R.T. Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr. Neuropharmacol. 2009, 7, 65–74. [Google Scholar] [CrossRef]
  121. Lasbleiz, C.; Peyrel, A.; Tarot, P.; Sarniguet, J.; Crouzier, L.; Cubedo, N.; Delprat, B.; Rossel, M.; Maurice, T.; Liévens, J.C. Sigma-1 receptor agonist PRE-084 confers protection against TAR DNA-binding protein-43 toxicity through NRF2 signalling. Redox. Biol. 2022, 58, 102542. [Google Scholar] [CrossRef]
  122. Estévez-Silva, H.M.; Cuesto, G.; Romero, N.; Brito-Armas, J.M.; Acevedo-Arozena, A.; Acebes, Á.; Marcellino, D.J. Pridopidine Promotes Synaptogenesis and Reduces Spatial Memory Deficits in the Alzheimer’s Disease APP/PS1 Mouse Model. Neurotherapeutics 2022, 19, 1566–1587. [Google Scholar] [CrossRef] [PubMed]
  123. Lahmy, V.; Long, R.; Morin, D.; Villard, V.; Maurice, T. Mitochondrial protection by the mixed muscarinic/σ1 ligand ANAVEX2-73, a tetrahydrofuran derivative, in Aβ25-35 peptide-injected mice, a nontransgenic Alzheimer’s disease model. Front. Cell. Neurosci. 2014, 8, 463. [Google Scholar] [CrossRef]
  124. Freyssin, A.; Carles, A.; Guehairia, S.; Rubinstenn, G.; Maurice, T. Fluoroethylnormemantine (FENM) shows synergistic protection in combination with a sigma-1 receptor agonist in a mouse model of Alzheimer’s disease. Neuropharmacology 2024, 242, 109733. [Google Scholar] [CrossRef] [PubMed]
  125. Christmann, U.; Garriga, L.; Llorente, A.V.; Diaz, J.L.; Pascual, R.; Bordas, M.; Dordal, A.; Porras, M.; Yeste, S.; Reinoso, R.F.; et al. WLB-87848, a Selective sigma(1) Receptor Agonist, with an Unusually Positioned NH Group as Positive Ionizable Moiety and Showing Neuroprotective Activity. J. Med. Chem. 2024, 67, 9150–9164. [Google Scholar] [CrossRef]
  126. Prasanth, M.I.; Verma, K.; Brimson, S.; Tencomnao, T.; Brimson, J.M. Simple ammonium salt and sigma-1 receptor ligand dipentylammonium provides neuroprotective effects in cell culture and Caenorhabditis elegans models of Alzheimer’s disease. Biomed. Pharmacother. 2024, 173, 116455. [Google Scholar] [CrossRef]
  127. Brimson, J.M.; Safrany, S.T.; Qassam, H.; Tencomnao, T. Dipentylammonium Binds to the Sigma-1 Receptor and Protects Against Glutamate Toxicity, Attenuates Dopamine Toxicity and Potentiates Neurite Outgrowth in Various Cultured Cell Lines. Neurotox. Res. 2018, 34, 263–272. [Google Scholar] [CrossRef]
  128. Bhattacharyya, R.; Black, S.E.; Lotlikar, M.S.; Fenn, R.H.; Jorfi, M.; Kovacs, D.M.; Tanzi, R.E. Axonal generation of amyloid-β from palmitoylated APP in mitochondria-associated endoplasmic reticulum membranes. Cell Rep. 2021, 35, 109134. [Google Scholar] [CrossRef]
  129. Rawat, P.; Sehar, U.; Bisht, J.; Selman, A.; Culberson, J.; Reddy, P.H. Phosphorylated Tau in Alzheimer’s Disease and Other Tauopathies. Int. J. Mol. Sci. 2022, 23, 12841. [Google Scholar] [CrossRef]
  130. Tsai, S.Y.; Pokrass, M.J.; Klauer, N.R.; Nohara, H.; Su, T.P. Sigma-1 receptor regulates Tau phosphorylation and axon extension by shaping p35 turnover via myristic acid. Proc. Natl. Acad. Sci. USA 2015, 112, 6742–6747. [Google Scholar] [CrossRef]
  131. Maurice, T.; Strehaiano, M.; Duhr, F.; Chevallier, N. Amyloid toxicity is enhanced after pharmacological or genetic invalidation of the σ(1) receptor. Behav. Brain Res. 2018, 339, 1–10. [Google Scholar] [CrossRef] [PubMed]
  132. Izzo, N.J.; Yuede, C.M.; LaBarbera, K.M.; Limegrover, C.S.; Rehak, C.; Yurko, R.; Waybright, L.; Look, G.; Rishton, G.; Safferstein, H.; et al. Preclinical and clinical biomarker studies of CT1812: A novel approach to Alzheimer’s disease modification. Alzheimer’s Dement. 2021, 17, 1365–1382. [Google Scholar] [CrossRef] [PubMed]
  133. Christmann, U.; Díaz, J.L.; Pascual, R.; Bordas, M.; Álvarez, I.; Monroy, X.; Porras, M.; Yeste, S.; Reinoso, R.F.; Merlos, M.; et al. Discovery of WLB-89462, a New Drug-like and Highly Selective σ(2) Receptor Ligand with Neuroprotective Properties. J. Med. Chem. 2023, 66, 12499–12519. [Google Scholar] [CrossRef] [PubMed]
  134. Yi, B.; Sahn, J.J.; Ardestani, P.M.; Evans, A.K.; Scott, L.L.; Chan, J.Z.; Iyer, S.; Crisp, A.; Zuniga, G.; Pierce, J.T.; et al. Small molecule modulator of sigma 2 receptor is neuroprotective and reduces cognitive deficits and neuroinflammation in experimental models of Alzheimer’s disease. J. Neurochem. 2017, 140, 561–575. [Google Scholar] [CrossRef]
  135. An, Y.; Qi, Y.; Li, Y.; Li, Z.; Yang, C.; Jia, D. Activation of the sigma-1 receptor attenuates blood-brain barrier disruption by inhibiting amyloid deposition in Alzheimer’s disease mice. Neurosci. Lett. 2022, 774, 136528. [Google Scholar] [CrossRef]
  136. Macfarlane, S.; Grimmer, T.; Teo, K.; O’Brien, T.J.; Woodward, M.; Grunfeld, J.; Mander, A.; Brodtmann, A.; Brew, B.J.; Morris, P.; et al. Blarcamesine for the treatment of Early Alzheimer’s Disease: Results from the ANAVEX2-73-AD-004 Phase IIB/III trial. J. Prev. Alzheimer’s Dis. 2025, 12, 100016. [Google Scholar] [CrossRef]
  137. Schneider, L.S.; Thomas, R.G.; Hendrix, S.; Rissman, R.A.; Brewer, J.B.; Salmon, D.P.; Oltersdorf, T.; Okuda, T.; Feldman, H.H.; Alzheimer’s Disease Cooperative Study, T.S.G. Safety and Efficacy of Edonerpic Maleate for Patients With Mild to Moderate Alzheimer Disease: A Phase 2 Randomized Clinical Trial. JAMA Neurol. 2019, 76, 1330–1339. [Google Scholar] [CrossRef]
  138. Kaur, G.; Dar, Z.A.; Bajpai, A.; Singh, R.; Bansal, R. Clinical Update on an Anti-Alzheimer Drug Candidate CT1812: A Sigma-2 Receptor Antagonist. Clin. Ther. 2024, 46, e21–e28. [Google Scholar] [CrossRef]
  139. Renton, A.E.; Majounie, E.; Waite, A.; Simon-Sanchez, J.; Rollinson, S.; Gibbs, J.R.; Schymick, J.C.; Laaksovirta, H.; van Swieten, J.C.; Myllykangas, L.; et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011, 72, 257–268. [Google Scholar] [CrossRef]
  140. Watanabe, S.; Ilieva, H.; Tamada, H.; Nomura, H.; Komine, O.; Endo, F.; Jin, S.; Mancias, P.; Kiyama, H.; Yamanaka, K. Mitochondria-associated membrane collapse is a common pathomechanism in SIGMAR1- and SOD1-linked ALS. EMBO Mol. Med. 2016, 8, 1421–1437. [Google Scholar] [CrossRef]
  141. Watanabe, S.; Horiuchi, M.; Murata, Y.; Komine, O.; Kawade, N.; Sobue, A.; Yamanaka, K. Sigma-1 receptor maintains ATAD3A as a monomer to inhibit mitochondrial fragmentation at the mitochondria-associated membrane in amyotrophic lateral sclerosis. Neurobiol. Dis. 2023, 179, 106031. [Google Scholar] [CrossRef] [PubMed]
  142. Chua, J.P.; De Calbiac, H.; Kabashi, E.; Barmada, S.J. Autophagy and ALS: Mechanistic insights and therapeutic implications. Autophagy 2022, 18, 254–282. [Google Scholar] [CrossRef] [PubMed]
  143. Lin, C.Y.; Wu, H.E.; Weng, E.F.; Wu, H.C.; Su, T.P.; Wang, S.M. Fluvoxamine Exerts Sigma-1R to Rescue Autophagy via Pom121-Mediated Nucleocytoplasmic Transport of TFEB. Mol. Neurobiol. 2024, 61, 5282–5294. [Google Scholar] [CrossRef] [PubMed]
  144. Estévez-Silva, H.M.; Mediavilla, T.; Giacobbo, B.L.; Liu, X.; Sultan, F.R.; Marcellino, D.J. Pridopidine modifies disease phenotype in a SOD1 mouse model of amyotrophic lateral sclerosis. Eur. J. Neurosci. 2022, 55, 1356–1372. [Google Scholar] [CrossRef]
  145. Gaja-Capdevila, N.; Hernández, N.; Navarro, X.; Herrando-Grabulosa, M. Sigma-1 Receptor is a Pharmacological Target to Promote Neuroprotection in the SOD1(G93A) ALS Mice. Front. Pharmacol. 2021, 12, 780588. [Google Scholar] [CrossRef]
  146. Zu, T.; Liu, Y.; Banez-Coronel, M.; Reid, T.; Pletnikova, O.; Lewis, J.; Miller, T.M.; Harms, M.B.; Falchook, A.E.; Subramony, S.H.; et al. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc. Natl. Acad. Sci. USA 2013, 110, E4968–E4977. [Google Scholar] [CrossRef]
  147. Lee, P.T.; Liévens, J.C.; Wang, S.M.; Chuang, J.Y.; Khalil, B.; Wu, H.E.; Chang, W.C.; Maurice, T.; Su, T.P. Sigma-1 receptor chaperones rescue nucleocytoplasmic transport deficit seen in cellular and Drosophila ALS/FTD models. Nat. Commun. 2020, 11, 5580. [Google Scholar] [CrossRef]
  148. Rouleau, G.A.; Clark, A.W.; Rooke, K.; Pramatarova, A.; Krizus, A.; Suchowersky, O.; Julien, J.P.; Figlewicz, D. SOD1 mutation is associated with accumulation of neurofilaments in amyotrophic lateral sclerosis. Ann. Neurol. 1996, 39, 128–131. [Google Scholar] [CrossRef] [PubMed]
  149. Ionescu, A.; Gradus, T.; Altman, T.; Maimon, R.; Saraf Avraham, N.; Geva, M.; Hayden, M.; Perlson, E. Targeting the Sigma-1 Receptor via Pridopidine Ameliorates Central Features of ALS Pathology in a SOD1(G93A) Model. Cell Death Dis. 2019, 10, 210. [Google Scholar] [CrossRef]
  150. Writing Committee for the, H.A.L.S.P.T.; Shefner, J.M.; Oskarsson, B.; Macklin, E.A.; Chibnik, L.B.; Quintana, M.; Saville, B.R.; Detry, M.A.; Vestrucci, M.; Marion, J.; et al. Pridopidine in Amyotrophic Lateral Sclerosis: The HEALEY ALS Platform Trial. JAMA 2025, 333, 1128–1137. [Google Scholar] [CrossRef]
  151. Martin, J.B.; Gusella, J.F. Huntington’s disease. Pathogenesis and management. N. Engl. J. Med. 1986, 315, 1267–1276. [Google Scholar] [CrossRef] [PubMed]
  152. Carmo, C.; Naia, L.; Lopes, C.; Rego, A.C. Mitochondrial Dysfunction in Huntington’s Disease. Adv. Exp. Med. Biol. 2018, 1049, 59–83. [Google Scholar] [CrossRef] [PubMed]
  153. Eddings, C.R.; Arbez, N.; Akimov, S.; Geva, M.; Hayden, M.R.; Ross, C.A. Pridopidine protects neurons from mutant-huntingtin toxicity via the sigma-1 receptor. Neurobiol. Dis. 2019, 129, 118–129. [Google Scholar] [CrossRef] [PubMed]
  154. Kusko, R.; Dreymann, J.; Ross, J.; Cha, Y.; Escalante-Chong, R.; Garcia-Miralles, M.; Tan, L.J.; Burczynski, M.E.; Zeskind, B.; Laifenfeld, D.; et al. Large-scale transcriptomic analysis reveals that pridopidine reverses aberrant gene expression and activates neuroprotective pathways in the YAC128 HD mouse. Mol. Neurodegener. 2018, 13, 25. [Google Scholar] [CrossRef]
  155. Kalathur, R.K.; Giner-Lamia, J.; Machado, S.; Barata, T.; Ayasolla, K.R.; Futschik, M.E. The unfolded protein response and its potential role in Huntington’s disease elucidated by a systems biology approach. F1000Research 2015, 4, 103. [Google Scholar] [CrossRef]
  156. Shenkman, M.; Geva, M.; Gershoni-Emek, N.; Hayden, M.R.; Lederkremer, G.Z. Pridopidine reduces mutant huntingtin-induced endoplasmic reticulum stress by modulation of the Sigma-1 receptor. J. Neurochem. 2021, 158, 467–481. [Google Scholar] [CrossRef]
  157. Bol’shakova, A.V.; Kraskovskaya, N.A.; Gainullina, A.N.; Kukanova, E.O.; Vlasova, O.L.; Bezprozvanny, I.B. Neuroprotective Effect of σ1-Receptors on the Cell Model of Huntington’s Disease. Bull. Exp. Biol. Med. 2017, 164, 252–258. [Google Scholar] [CrossRef]
  158. Ryskamp, D.; Wu, J.; Geva, M.; Kusko, R.; Grossman, I.; Hayden, M.; Bezprozvanny, I. The sigma-1 receptor mediates the beneficial effects of pridopidine in a mouse model of Huntington disease. Neurobiol. Dis. 2017, 97, 46–59. [Google Scholar] [CrossRef]
  159. McGarry, A.; Auinger, P.; Kieburtz, K.; Geva, M.; Mehra, M.; Abler, V.; Grachev, I.D.; Gordon, M.F.; Savola, J.M.; Gandhi, S.; et al. Additional Safety and Exploratory Efficacy Data at 48 and 60 Months from Open-HART, an Open-Label Extension Study of Pridopidine in Huntington Disease. J. Huntingt. Dis. 2020, 9, 173–184. [Google Scholar] [CrossRef]
  160. Goldberg, Y.P.; Navon-Perry, L.; Cruz-Herranz, A.; Chen, K.; Hecker-Barth, G.; Spiegel, K.; Cohen, Y.; Niethammer, M.; Tan, A.M.; Schuring, H.; et al. The Safety Profile of Pridopidine, a Novel Sigma-1 Receptor Agonist for the Treatment of Huntington’s Disease. CNS Drugs 2025, 39, 485–498. [Google Scholar] [CrossRef]
  161. Alexoudi, A.; Alexoudi, I.; Gatzonis, S. Parkinson’s disease pathogenesis, evolution and alternative pathways: A review. Rev. Neurol. 2018, 174, 699–704. [Google Scholar] [CrossRef] [PubMed]
  162. Calabresi, P.; Mechelli, A.; Natale, G.; Volpicelli-Daley, L.; Di Lazzaro, G.; Ghiglieri, V. Alpha-synuclein in Parkinson’s disease and other synucleinopathies: From overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis. 2023, 14, 176. [Google Scholar] [CrossRef] [PubMed]
  163. Hong, J.; Wang, L.; Zhang, T.; Zhang, B.; Chen, L. Sigma-1 receptor knockout increases α-synuclein aggregation and phosphorylation with loss of dopaminergic neurons in substantia nigra. Neurobiol. Aging 2017, 59, 171–183. [Google Scholar] [CrossRef] [PubMed]
  164. Limegrover, C.S.; Yurko, R.; Izzo, N.J.; LaBarbera, K.M.; Rehak, C.; Look, G.; Rishton, G.; Safferstein, H.; Catalano, S.M. Sigma-2 receptor antagonists rescue neuronal dysfunction induced by Parkinson’s patient brain-derived α-synuclein. J. Neurosci. Res. 2021, 99, 1161–1176. [Google Scholar] [CrossRef]
  165. Vetel, S.; Foucault-Fruchard, L.; Tronel, C.; Buron, F.; Vergote, J.; Bodard, S.; Routier, S.; Sérrière, S.; Chalon, S. Neuroprotective and anti-inflammatory effects of a therapy combining agonists of nicotinic α7 and σ1 receptors in a rat model of Parkinson’s disease. Neural Regen. Res. 2021, 16, 1099–1104. [Google Scholar] [CrossRef]
  166. Wang, M.; Wan, C.; He, T.; Han, C.; Zhu, K.; Waddington, J.L.; Zhen, X. Sigma-1 receptor regulates mitophagy in dopaminergic neurons and contributes to dopaminergic protection. Neuropharmacology 2021, 196, 108360. [Google Scholar] [CrossRef]
  167. Francardo, V.; Geva, M.; Bez, F.; Denis, Q.; Steiner, L.; Hayden, M.R.; Cenci, M.A. Pridopidine Induces Functional Neurorestoration Via the Sigma-1 Receptor in a Mouse Model of Parkinson’s Disease. Neurotherapeutics 2019, 16, 465–479. [Google Scholar] [CrossRef]
  168. Pandey, S.; Srivanitchapoom, P. Levodopa-induced Dyskinesia: Clinical Features, Pathophysiology, and Medical Management. Ann. Indian Acad. Neurol. 2017, 20, 190–198. [Google Scholar] [CrossRef]
  169. Johnston, T.H.; Geva, M.; Steiner, L.; Orbach, A.; Papapetropoulos, S.; Savola, J.M.; Reynolds, I.J.; Ravenscroft, P.; Hill, M.; Fox, S.H.; et al. Pridopidine, a clinic-ready compound, reduces 3,4-dihydroxyphenylalanine-induced dyskinesia in Parkinsonian macaques. Mov. Disord. 2019, 34, 708–716. [Google Scholar] [CrossRef]
  170. Fox, S.H.; Metman, L.V.; Nutt, J.G.; Brodsky, M.; Factor, S.A.; Lang, A.E.; Pope, L.E.; Knowles, N.; Siffert, J. Trial of dextromethorphan/quinidine to treat levodopa-induced dyskinesia in Parkinson’s disease. Mov. Disord. 2017, 32, 893–903. [Google Scholar] [CrossRef]
  171. Sun, Y.; Sukumaran, P.; Singh, B.B. Sigma1 Receptor Inhibits TRPC1-Mediated Ca(2+) Entry That Promotes Dopaminergic Cell Death. Cell. Mol. Neurobiol. 2021, 41, 1245–1255. [Google Scholar] [CrossRef] [PubMed]
  172. Hong, J.; Sha, S.; Zhou, L.; Wang, C.; Yin, J.; Chen, L. Sigma-1 receptor deficiency reduces MPTP-induced parkinsonism and death of dopaminergic neurons. Cell Death Dis. 2015, 6, e1832. [Google Scholar] [CrossRef]
  173. Suzuki, K.; Suzuki, Y. Globoid cell leucodystrophy (Krabbe’s disease): Deficiency of galactocerebroside beta-galactosidase. Proc. Natl. Acad. Sci. USA 1970, 66, 302–309. [Google Scholar] [CrossRef]
  174. Papakyriakopoulou, P.; Valsami, G.; Dev, K.K. The Effect of Donepezil Hydrochloride in the Twitcher Mouse Model of Krabbe Disease. Mol. Neurobiol. 2024, 61, 8688–8701. [Google Scholar] [CrossRef] [PubMed]
  175. Barrett, T.G.; Bundey, S.E. Wolfram (DIDMOAD) syndrome. J. Med. Genet. 1997, 34, 838–841. [Google Scholar] [CrossRef]
  176. Fischer, T.T.; Nguyen, L.D.; Ehrlich, B.E. Boosting ER-mitochondria calcium transfer to treat Wolfram syndrome. Cell Calcium 2022, 104, 102572. [Google Scholar] [CrossRef]
  177. Crouzier, L.; Danese, A.; Yasui, Y.; Richard, E.M.; Liévens, J.C.; Patergnani, S.; Couly, S.; Diez, C.; Denus, M.; Cubedo, N.; et al. Activation of the sigma-1 receptor chaperone alleviates symptoms of Wolfram syndrome in preclinical models. Sci. Transl. Med. 2022, 14, eabh3763. [Google Scholar] [CrossRef] [PubMed]
  178. Atzmon, A.; Herrero, M.; Sharet-Eshed, R.; Gilad, Y.; Senderowitz, H.; Elroy-Stein, O. Drug Screening Identifies Sigma-1-Receptor as a Target for the Therapy of VWM Leukodystrophy. Front. Mol. Neurosci. 2018, 11, 336. [Google Scholar] [CrossRef]
  179. Noyes, K.; Weinstock-Guttman, B. Impact of diagnosis and early treatment on the course of multiple sclerosis. Am. J. Manag. Care 2013, 19, s321–s331. [Google Scholar]
  180. Collina, S.; Rui, M.; Stotani, S.; Bignardi, E.; Rossi, D.; Curti, D.; Giordanetto, F.; Malacrida, A.; Scuteri, A.; Cavaletti, G. Are sigma receptor modulators a weapon against multiple sclerosis disease? Future Med. Chem. 2017, 9, 2029–2051. [Google Scholar] [CrossRef]
  181. Behensky, A.A.; Katnik, C.; Yin, H.; Cuevas, J. Activation of Sigma Receptors with Afobazole Modulates Microglial, but Not Neuronal, Apoptotic Gene Expression in Response to Long-Term Ischemia Exposure. Front. Neurosci. 2019, 13, 414. [Google Scholar] [CrossRef] [PubMed]
  182. Naaz, S.; Ozair, E. Dexmedetomidine in current anaesthesia practice- a review. J. Clin. Diagn. Res. 2014, 8, GE01-04. [Google Scholar] [CrossRef] [PubMed]
  183. Kenche, H.; Singh, M.; Smith, J.; Shen, K. Neuronal mitochondrial dysfunction in a cellular model of circadian rhythm disruption is rescued by donepezil. Biochem. Biophys. Res. Commun. 2021, 567, 56–62. [Google Scholar] [CrossRef] [PubMed]
  184. Birks, J.S.; Harvey, R.J. Donepezil for dementia due to Alzheimer’s disease. Cochrane Database Syst. Rev. 2018, 6, CD001190. [Google Scholar] [CrossRef]
  185. Xu, Q.; Ji, X.F.; Chi, T.Y.; Liu, P.; Jin, G.; Chen, L.; Zou, L.B. Sigma-1 receptor in brain ischemia/reperfusion: Possible role in the NR2A-induced pathway to regulate brain-derived neurotrophic factor. J. Neurol. Sci. 2017, 376, 166–175. [Google Scholar] [CrossRef]
  186. Ryskamp, D.; Wu, L.; Wu, J.; Kim, D.; Rammes, G.; Geva, M.; Hayden, M.; Bezprozvanny, I. Pridopidine stabilizes mushroom spines in mouse models of Alzheimer’s disease by acting on the sigma-1 receptor. Neurobiol. Dis. 2019, 124, 489–504. [Google Scholar] [CrossRef]
  187. Kimura, T.; Hong Nguyen, P.T.; Ho, S.A.; Tran, A.H.; Ono, T.; Nishijo, H. T-817MA, a neurotrophic agent, ameliorates the deficits in adult neurogenesis and spatial memory in rats infused i.c.v. with amyloid-beta peptide. Br. J. Pharmacol. 2009, 157, 451–463. [Google Scholar] [CrossRef]
  188. Lizama, B.N.; North, H.A.; Pandey, K.; Williams, C.; Duong, D.; Cho, E.; Di Caro, V.; Ping, L.; Blennow, K.; Zetterberg, H.; et al. An interim exploratory proteomics biomarker analysis of a phase 2 clinical trial to assess the impact of CT1812 in Alzheimer’s disease. Neurobiol. Dis. 2024, 199, 106575. [Google Scholar] [CrossRef]
  189. Grundman, M.; Morgan, R.; Lickliter, J.D.; Schneider, L.S.; DeKosky, S.; Izzo, N.J.; Guttendorf, R.; Higgin, M.; Pribyl, J.; Mozzoni, K.; et al. A phase 1 clinical trial of the sigma-2 receptor complex allosteric antagonist CT1812, a novel therapeutic candidate for Alzheimer’s disease. Alzheimer’s Dement. 2019, 5, 20–26. [Google Scholar] [CrossRef]
  190. Vijverberg, E.; de Haan, W.; Scheijbeler, E.; Hamby, M.E.; Catalano, S.; Scheltens, P.; Grundman, M.; Caggiano, A.O. A Pilot Electroencephalography Study of the Effect of CT1812 Treatment on Synaptic Activity in Patients with Mild to Moderate Alzheimer’s Disease. J. Prev. Alzheimer’s Dis. 2024, 11, 1809–1817. [Google Scholar] [CrossRef]
Figure 1. Structural framework of the review: from S1R biology to therapeutic application in neurological disease. Abbreviations: S1R, Sigma-1 receptor; TBI, traumatic brain injury; ALS, amyotrophic lateral sclerosis.
Figure 1. Structural framework of the review: from S1R biology to therapeutic application in neurological disease. Abbreviations: S1R, Sigma-1 receptor; TBI, traumatic brain injury; ALS, amyotrophic lateral sclerosis.
Biomedicines 13 01409 g001
Figure 2. S1R localization, activation, and downstream interactions. Under basal conditions, S1R resides at the mitochondria-associated endoplasmic reticulum membrane (MAM), tethered to BiP. Upon ER stress, S1R dissociates from BiP and binds to inositol 1,4,5-triphosphate receptors, facilitating calcium signaling between the ER and mitochondria. Activated S1R translocates to other compartments, such as the plasma membrane. S1R can modulate ion channels, NMDA receptors, and G-protein coupled receptors. These downstream interactions mediate calcium homeostasis as well as reductions in ER dysfunction and oxidative stress. S1R is implicated in therapeutic benefits for the brain, heart, liver, and kidney. Abbreviations: S1R, Sigma-1 receptor; MAM, mitochondria-associated membrane; BiP, binding immunoglobulin protein; ER, endoplasmic reticulum; IP3, inositol 1,4,5-triphosphate; NMDA, N-methyl-D-aspartate. Figure created using Servier Medical Art (CC BY 3.0 license).
Figure 2. S1R localization, activation, and downstream interactions. Under basal conditions, S1R resides at the mitochondria-associated endoplasmic reticulum membrane (MAM), tethered to BiP. Upon ER stress, S1R dissociates from BiP and binds to inositol 1,4,5-triphosphate receptors, facilitating calcium signaling between the ER and mitochondria. Activated S1R translocates to other compartments, such as the plasma membrane. S1R can modulate ion channels, NMDA receptors, and G-protein coupled receptors. These downstream interactions mediate calcium homeostasis as well as reductions in ER dysfunction and oxidative stress. S1R is implicated in therapeutic benefits for the brain, heart, liver, and kidney. Abbreviations: S1R, Sigma-1 receptor; MAM, mitochondria-associated membrane; BiP, binding immunoglobulin protein; ER, endoplasmic reticulum; IP3, inositol 1,4,5-triphosphate; NMDA, N-methyl-D-aspartate. Figure created using Servier Medical Art (CC BY 3.0 license).
Biomedicines 13 01409 g002
Figure 3. Role of S1R in nervous system injury. Events such as ischemic stroke and TBI trigger a cascade of processes such as BBB breakdown, microglial activation, and neuronal apoptosis. These are mediated through ER stress, spreading depolarizations, and neuroinflammation. Activation of S1R reduces these mechanisms and promotes neuroprotection. Several S1R modulators have been studied in preclinical models and demonstrate beneficial effects. Abbreviations: S1R, Sigma-1 receptor; TBI, traumatic brain injury; BBB, blood–brain barrier; ER, endoplasmic reticulum; IL1-β, interleukin-1 beta; IL-6, interleukin-6; TNF-α, tumor necrosis factor alpha; p-PERK, phosphorylated protein kinase RNA-like ER kinase; p-IRE1α, phosphorylated inositol-requiring enzyme 1 alpha; JNK, c-Jun N-terminal kinase; BDNF, brain-derived neurotrophic factor; CNPase, 2′,3′-cyclic-nucleotide 3′-phosphodiesterase; MOG, myelin oligodendrocyte glycoprotein; PDGFRα, platelet-derived growth factor receptor alpha. Figure created using Servier Medical Art (CC BY 3.0 license).
Figure 3. Role of S1R in nervous system injury. Events such as ischemic stroke and TBI trigger a cascade of processes such as BBB breakdown, microglial activation, and neuronal apoptosis. These are mediated through ER stress, spreading depolarizations, and neuroinflammation. Activation of S1R reduces these mechanisms and promotes neuroprotection. Several S1R modulators have been studied in preclinical models and demonstrate beneficial effects. Abbreviations: S1R, Sigma-1 receptor; TBI, traumatic brain injury; BBB, blood–brain barrier; ER, endoplasmic reticulum; IL1-β, interleukin-1 beta; IL-6, interleukin-6; TNF-α, tumor necrosis factor alpha; p-PERK, phosphorylated protein kinase RNA-like ER kinase; p-IRE1α, phosphorylated inositol-requiring enzyme 1 alpha; JNK, c-Jun N-terminal kinase; BDNF, brain-derived neurotrophic factor; CNPase, 2′,3′-cyclic-nucleotide 3′-phosphodiesterase; MOG, myelin oligodendrocyte glycoprotein; PDGFRα, platelet-derived growth factor receptor alpha. Figure created using Servier Medical Art (CC BY 3.0 license).
Biomedicines 13 01409 g003
Figure 4. Proposed protective mechanisms and pharmaceutical implications of sigma-1 receptor in Alzheimer’s disease. This schematic illustrates the progression of AD driven by the deposition of amyloid plaques in the nervous system. S1R activation modulates several key processes in AD pathogenesis to slow cognitive decline and disease progression. Abbreviations: S1R, Sigma-1 receptor; AD, Alzheimer’s disease; NFkB, nuclear factor kappa B; TNFα, tumor necrosis factor alpha; IL-6, interleukin-6; Bax, Bcl-2-associated X protein; MAM, mitochondria-associated membrane; ERK, extracellular signal-regulated kinase; Akt, protein kinase B. Figure created using Servier Medical Art (CC BY 3.0 license).
Figure 4. Proposed protective mechanisms and pharmaceutical implications of sigma-1 receptor in Alzheimer’s disease. This schematic illustrates the progression of AD driven by the deposition of amyloid plaques in the nervous system. S1R activation modulates several key processes in AD pathogenesis to slow cognitive decline and disease progression. Abbreviations: S1R, Sigma-1 receptor; AD, Alzheimer’s disease; NFkB, nuclear factor kappa B; TNFα, tumor necrosis factor alpha; IL-6, interleukin-6; Bax, Bcl-2-associated X protein; MAM, mitochondria-associated membrane; ERK, extracellular signal-regulated kinase; Akt, protein kinase B. Figure created using Servier Medical Art (CC BY 3.0 license).
Biomedicines 13 01409 g004
Figure 5. Disease-specific pathways of Sigma-1 receptor activation in various neurodegenerative diseases. In PD, S1R activation protects dopaminergic neurons and counteracts alpha-synuclein accumulation. In ALS, S1R reduces SOD1 and RAN accumulation, preventing the loss of motor neurons. In HD, S1R helps prevent degeneration of the striatum by relieving ER stress and stimulating neurotrophic proteins. Abbreviations: S1R, Sigma-1 receptor; GDNF, glial cell-derived neurotrophic factor; BDNF, brain-derived neurotrophic factor; ERK, extracellular signal-regulated kinase; GR, glucocorticoid receptor; D1R, dopamine D1 receptor; cAMP, cyclic adenosine monophosphate; TFEB, transcription factor EB; RAN, repeat-associated non-AUG. Figure created using Servier Medical Art (CC BY 3.0 license).
Figure 5. Disease-specific pathways of Sigma-1 receptor activation in various neurodegenerative diseases. In PD, S1R activation protects dopaminergic neurons and counteracts alpha-synuclein accumulation. In ALS, S1R reduces SOD1 and RAN accumulation, preventing the loss of motor neurons. In HD, S1R helps prevent degeneration of the striatum by relieving ER stress and stimulating neurotrophic proteins. Abbreviations: S1R, Sigma-1 receptor; GDNF, glial cell-derived neurotrophic factor; BDNF, brain-derived neurotrophic factor; ERK, extracellular signal-regulated kinase; GR, glucocorticoid receptor; D1R, dopamine D1 receptor; cAMP, cyclic adenosine monophosphate; TFEB, transcription factor EB; RAN, repeat-associated non-AUG. Figure created using Servier Medical Art (CC BY 3.0 license).
Biomedicines 13 01409 g005
Table 1. Summary of key S1R and S2R ligands with indications, mechanisms, and developmental status.
Table 1. Summary of key S1R and S2R ligands with indications, mechanisms, and developmental status.
LigandSigma Receptor TargetDiseaseMechanism of ActionDevelopmental StatusRef.
AF710BSigma 1 AgonistAlzheimer’s Disease↓ cognitive impairment
↓ amyloid plaques
↓ inflammatory cytokines
Preclinical[108,118]
Aniline DerivativesSigma 1 AgonistStroke↓ p-PERK and p-IRE1α expression
↓ ER stress
Preclinical[49]
AfobazoleSigma 1 AgonistStroke↓ Bax and caspase-3
↑ Bcl-2
↓ neuronal death
↓ reactive astrocytes
Preclinical[55,181]
BlarcamesineSigma 1 AgonistAlzheimer’s Disease, Multiple SclerosisMS:
↑ oligodendrogliosis
↓ apoptosis and excitotoxicity
↓ reactive oxygen species and quinolinic acid
Phase IIb/III (Alzheimer’s Disease)
Preclinical (Multiple Sclerosis)
[90,136]
DexmedetomidineSigma 1 AgonistStroke↓ BBB permeability
↓ neuronal damage
↓ CHOP, Caspase-3, and JNK
↑ Occludin stabilization
Approved (Sedation)[48,67,182]
DipentylammoniumSigma 1 AgonistAlzheimer’s Disease↑ neurite growth length
↓ excitotoxicity
↓ NFkB activation
Preclinical[126,127]
DonepezilSigma 1 AgonistGeneral Neurodegeneration↑ oxidative respiration
↑ mitochondrial membrane potentials
Approved (Alzheimer’s Disease)[183,184]
FluvoxamineSigma 1 AgonistALS↑ chaperone activity
↑ Pom121 expression
↑ TFEB nuclear translocation
↑ LC3-II expression
Preclinical[143]
N,N-DimethyltryptamineSigma 1 AgonistAlzheimer’s Disease, StrokeAlzheimer’s Disease:
↓ cognitive impairment
↓ amyloid plaques
↑ MAM-associated proteins
Stroke:
↓ apoptosis and ferroptosis
↓ infarct size
↑ BDNF expression
↓ TNF-α, IL1-β, IL-6
Phase I (Alzheimer’s Disease)
Preclinical (Stroke)
[47,53,72,114]
OxeladinSigma 1 AgonistStroke↑ neurologic function
infarct size
↑ BDNF expression
Preclinical[52]
OZP002Sigma 1 AgonistAlzheimer’s Disease↓ cognitive impairment
↓ reactive oxygen species and lipid peroxidation
↓ Bax, TNFα, IL-6
↓ reactive gliosis
↑ synaptophysin and choline acetyltransferase
Preclinical[117]
(+)-PentazocineSigma 1 AgonistGeneral Neurodegeneration↓ microglial recruitment
↓ GAD, SOD, and p65
Preclinical[111]
PRE-084Sigma 1 AgonistALS, Alzheimer’s Disease, Parkinson’s Disease, StrokeALS:
↑ neuromuscular function
↑ BiP, EIF2α/ATF4, NRF2
Alzheimer’s Disease:
↑ hippocampal proliferation
↓ reactive gliosis
↓ microglial activation
Parkinson’s Disease:
↑ dopaminergic protection
↓ microglial activation
↓ MPTP-induced damage
Stroke:
↓ spreading depolarizations
↑ myelin density
↑ CNpase, MOG, NG2 cells, PDGFRα
↓ BBB permeability
↓ learning impairments
↑ BDNF, NR2A, CaMKIV, and TORC1
Preclinical[58,62,72,116,121,145,165,166,185]
PridopidineSigma 1 AgonistALS, Alzheimer’s Disease, Parkinson’s Disease, Huntington’s DiseaseALS:
↑ neuromuscular function
↑ ERK
↓ SOD1 aggregation
Alzheimer’s Disease:
↓ excitotoxicity
↑ synapses and dendritic spines
↑ ERK and Akt signaling
↑ long term potentiation
Parkinson’s Disease:
↑ dopaminergic protection
↑ GDNF, BDNF, ERK
Huntington’s Disease:
↑ BDNF, TrkB, GR, D1R, cAMP
↓ ER stress
↓ reactive oxygen species
↑ calbindin and homer1a
Phase III (Huntington’s Disease)
Phase II/III (ALS)
Preclinical (Alzheimer’s Disease, Parkinson’s Disease)
[89,94,122,144,149,154,156,158,160,167,169,186]
T-817MASigma 1 AgonistAlzheimer’s Disease↓ cognitive impairment
↑ hippocampal proliferation
Phase IIa[137,187]
TS-157Sigma 1 AgonistStroke↑ neurite outgrowth
↑ ERK signaling
↑ motor recovery
Preclinical[64]
UlinastatinSigma 1 AgonistStroke↑ motor recoveryPreclinical[63]
WLB-87848Sigma 1 AgonistAlzheimer’s Disease↓ cognitive impairment
↑ neuron viability
Preclinical[125]
BD1063Sigma 1 AntagonistALS↑ neuromuscular functionPreclinical[145]
WLB-89462Sigma 2 AgonistAlzheimer’s Disease↓ cognitive impairmentPreclinical[133]
CT1812Sigma 2 AntagonistAlzheimer’s Disease↓ cognitive impairment
↓ amyloid plaques
↓ phosphorylated tau fragments
Phase II[132,138,188,189,190]
S1RASigma 2 AntagonistStroke↑ stroke recovery
↓ reactive gliosis
↓ MMP-9 expression
Preclinical[57]
SAS-0132Sigma 2 AntagonistAlzheimer’s Disease↓ cognitive impairmentPreclinical[134]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Drewes, N.; Fang, X.; Gupta, N.; Nie, D. Pharmacological and Pathological Implications of Sigma-1 Receptor in Neurodegenerative Diseases. Biomedicines 2025, 13, 1409. https://doi.org/10.3390/biomedicines13061409

AMA Style

Drewes N, Fang X, Gupta N, Nie D. Pharmacological and Pathological Implications of Sigma-1 Receptor in Neurodegenerative Diseases. Biomedicines. 2025; 13(6):1409. https://doi.org/10.3390/biomedicines13061409

Chicago/Turabian Style

Drewes, Noah, Xiangwei Fang, Nikhil Gupta, and Daotai Nie. 2025. "Pharmacological and Pathological Implications of Sigma-1 Receptor in Neurodegenerative Diseases" Biomedicines 13, no. 6: 1409. https://doi.org/10.3390/biomedicines13061409

APA Style

Drewes, N., Fang, X., Gupta, N., & Nie, D. (2025). Pharmacological and Pathological Implications of Sigma-1 Receptor in Neurodegenerative Diseases. Biomedicines, 13(6), 1409. https://doi.org/10.3390/biomedicines13061409

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

Article Metrics

Back to TopTop