Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Neurosci., 19 December 2025

Sec. Neurodegeneration

Volume 19 - 2025 | https://doi.org/10.3389/fnins.2025.1733659

This article is part of the Research TopicInter-organelle membrane contacts in health and disease of the nervous systemView all articles

Sigma receptors and mitochondria-associated ER membranes are converging therapeutic targets for Alzheimer’s disease

  • Genetics and Aging Research Unit, Mass General Institute for Neurodegenerative Disease, Henry and Allison McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States

Alzheimer’s disease (AD) begins decades before clinical symptoms emerge. The “amyloid hypothesis” suggests that amyloid-β (Aβ) deposition initiates a cascade of tau hyperphosphorylation, neuroinflammation, and neuronal loss leading to cognitive decline. The recent success of anti-Aβ therapies such as Leqembi in prodromal or mild cognitive impaired patients underscores the importance of early intervention and Aβ clearance. However, safety and cost limitations highlight the need for alternative therapeutic strategies. Small-molecule modulators of Sigma-1 and Sigma-2 receptors (σ1R and σ2R) have emerged as promising candidates for AD treatment. σ1R agonists exhibit neuroprotective and anti-amnestic effects under pathological conditions without affecting normal cognition. Beyond AD, σ1R is implicated in several neurodegenerative diseases including ALS (amyotrophic lateral sclerosis), Parkinson’s, and Huntington’s diseases, stroke, and epilepsy. σ1R plays a key role at mitochondria-associated ER membranes (MAMs)—specialized lipid raft-like domains that form functional membrane contact sites between the endoplasmic reticulum (ER) and mitochondria. β-secretase (BACE1), γ-secretase, and their substrates APP and palmitoylated APP (palAPP) localize in the MAMs, promoting amyloidogenic Aβ production. MAMs serve as dynamic hubs for inter-organelle communication, calcium signaling, and lipid metabolism. The “MAM hypothesis” proposes that MAM dysregulation drives early AD pathology and persists throughout disease progression, contributing to neurofibrillary tangle formation, calcium imbalance, and neuroinflammation. This review aims to summarize the current understanding of σ1R-mediated regulation of MAMs and its neuroprotective mechanisms, highlighting potential therapeutic opportunities for targeting σ1R in AD and other neurodegenerative disorders.

Introduction

Alzheimer’s Disease (AD) pathophysiology begins decades before symptoms appear. Translational research, including genetic, biological, and biomarker studies, has significantly advanced our understanding of the “amyloid hypothesis” that proposes that the pathogenesis of is primarily caused by the deposition of amyloid-β (Aβ), which triggers tau phosphorylation, neuroinflammation, and neurodegeneration in the brain leading to cognitive impairment and memory deficit (Hampel et al., 2021). The recent success of anti-Aβ therapies (e.g., Leqembi) in patients with prodromal or mild cognitive impairment underscores the importance of Aβ clearance and the value of treating patients before symptoms appear (van Dyck et al., 2023; Budd Haeberlein et al., 2022). However, safety concerns and high costs highlight the need for alternative strategies.

Small-molecule therapies targeting Sigma-1 and -2 Receptors (σ1R and σ2R, respectively), offer a promising approach to treat AD (De Vos et al., 2012; Magalhaes Rebelo et al., 2020). σ1R agonists with favorable pharmacological profiles exhibit anti-amnestic effects in pathological conditions but not normal memory (Magalhaes Rebelo et al., 2020; Weber and Wünsch, 2017; Sałaciak and Pytka, 2022; Uchida et al., 2005; Maruszak et al., 2007; Huang et al., 2011; Feher et al., 2012; Wang and Jia, 2023; Fallica et al., 2021). σ1R is emerging as a unique therapeutic target for several neurodegenerative diseases including Amyotrophic Lateral Sclerosis (ALS), Parkinson’s, Huntington’s, and Alzheimer’s Diseases (PD, HD, and AD, respectively), stroke, and Epilepsy (Piechal et al., 2021; Malar et al., 2023). The precise mechanism underlying the neuroprotective regulatory functions of σ1R ligands also remains unclear.

σ1R is an endoplasmic reticulum (ER)-resident chaperone that under physiological conditions, binds with the chaperone-binding immunoglobulin protein (BiP)/glucose-regulated protein 78 (GRP78) on the ER. Upon activation, σ1R dissociates from BiP/GRP78 and interacts with type 3 inositol 1,4,5-trisphosphate receptor (IP3R3; Weng et al., 2017). IP3R3s then forms a tripartite complex with mitochondrial voltage-dependent anion-selective channel 1 (VDAC1) and glucose-regulated protein 75 (GRP75). This stabilizes the membrane contact sites (MCS) formed between the lipid raft (LR)-like dynamic membrane on the ER and the outer membrane of mitochondria (OMM), biochemically isolated as mitochondria-associated ER membranes or MAMs (Bhattacharyya et al., 2021; Fujimoto and Hayashi, 2011).

MAMs are specialized intracellular sites where β-secretase (BACE1) is found accumulating with its substrates APP (β-amyloid precursor protein) or palmitoylated APP (palAPP), along with γ-secretases producing neurotoxic Aβ are found (Bhattacharyya et al., 2021; Pera et al., 2017; Area-Gomez et al., 2009; Bhattacharyya et al., 2013). LR- or MAM-bound palAPP is considered a potential therapeutic target for AD because it serves as a preferential substrate for BACE1, the rate limiting step for Aβ generation in vitro and in vivo (reviewed in Wlodarczyk et al., 2024). MAMs are dynamic and multifunctional scaffold to enable crosstalk between the ER and mitochondria (Bhattacharyya et al., 2021; Zellmer et al., 2024). Recently coined “MAM hypothesis” proposes that MAMs play a critical role in Aβ production to initiate the pathogenic cascade of AD, including NFT formation, calcium dyshomeostasis, and neuroinflammation (Area-Gomez and Schon, 2017; Schon and Area-Gomez, 2013). In addition to AD, MAM perturbation has also been implicated in ALS, PD, and HD (Prinz et al., 2020; Voeltz et al., 2024).

The purpose of this review is to provide a comprehensive overview of the current understanding of σ1R’s role in neurodegeneration and neuroprotection. We will discuss its molecular mechanisms, interactions with key cellular pathways, and potential therapeutic implications. By elucidating the mechanistic insights into σ1R function, we aim to highlight future directions for research and therapeutic development in neurodegenerative disorders.

Targeting SIGMA-1 receptor (σ1R) in AD

The Sigma-1 receptor (σ1R), an endoplasmic reticulum (ER)-resident chaperone, is highly expressed in the brain and a promising target for neurodegenerative diseases, including AD (Piechal et al., 2021). σ1R is an atypical type I transmembrane (TM) protein that has no second messenger system (Schmidt et al., 2016). While the cellular stress such as the ER or oxidative stress triggers σ1R function, σ1R has no well-defined endogenous ligands except steroids, such as progesterone, testosterone, and neurosteroids that act as modifiers (Hayashi, 2019; Morales-Lázaro et al., 2019). Most σ1R ligands are synthetic small molecules. Several σ1R ligands show anti-amnestic effects in preclinical models and favorable pharmacokinetics (Sałaciak and Pytka, 2022; Wang and Jia, 2023). Notably, σ1R expression increases in early AD—likely as an adaptive response to cellular stress—and σ1R polymorphisms are linked to AD and other neurodegenerative conditions, and polymorphism in the σ1R gene is found to be associated with ALS/FTD (Frontotemporal Dementia), AD, and other neurodegenerative diseases (Uchida et al., 2005; Maruszak et al., 2007; Huang et al., 2011; Feher et al., 2012; Piechal et al., 2021; Luty et al., 2010; Al-Saif et al., 2011; Gregianin et al., 2016; Watanabe et al., 2016; Feher et al., 2012; Jin et al., 2015). σ1R interacts with multiple protein complexes to modulate several signaling pathways such as, cellular calcium homeostasis, excitotoxicity, ER stress, mitochondrial function. These pathways are critical to maintain cellular health and survival. This makes the σ1R an interesting target for the development of drugs for neurological diseases where neuronal loss or degeneration is a key part of the pathology. A postmortem study reported that σ1R levels decreased in hippocampus CA1 region of AD patients (Jansen et al., 1993). Direct visualization of σ1R in live mouse brains using a novel (11C)-labeled positron emission tomography (PET) probe, [11C]CNY-01, combined with immunocytochemistry (ICC) showed reduced σ1R levels in 5XFAD mouse brains compared to the WT, with σ1R reduction linked to amyloid and neuroinflammation pathologies (Bai et al., 2025). An earlier study using the PET probe [11C]SA4503 for σ1R in human brains of patients with early AD found diminished expression levels of σ1R compared to cognitively normal controls (Venkataraman et al., 2022). These findings add to the weight of evidence that the σ1R is important and that σ1R ligands may be of benefit to AD patients at their early stages. Regarding therapeutic potential, many σ1R agonists are FDA-approved and show promising anti-amnestic effects in pre-clinical studies of mild-to-moderate AD (Sałaciak and Pytka, 2022; Uchida et al., 2005; Maruszak et al., 2007; Huang et al., 2011; Feher et al., 2012; Wang and Jia, 2023; Rountree et al., 2013; Hampel et al., 2020). Recently, ANAVEX2-73 (blarcamesine), a mixed sigma-1 and muscarinic receptor ligand, has reached phase 2b/3 trials for AD (Hampel et al., 2020; Macfarlane et al., 2025). σ1R-agonists are gaining attention because they act as anti-amnestic agents only in pathological conditions but not in normal memory (reviewed in Sałaciak and Pytka, 2022).

While σ1R agonists show anti-amnestic effects, several prototypic σ1R agonists including PRE-084, (+)-pentazocine, (+)-SKF10047, 4-(N-benzylpiperidin-4-yl)-4-iodobenzamide (4-IBP), and SA4503 are reported to promote cancer cell proliferation and tumor growth, while some of these putative agonists inhibit cell proliferation and trigger cell cycle arrest and a few appear to have no effect on cell proliferation and tumor growth (Megalizzi et al., 2009; Megalizzi et al., 2007; Kim and Maher, 2017). These contradictory findings may be because the notion of agonist and antagonist classifications may be misleading for σ1R because it is an atypical type I transmembrane (TM) protein that has no second messenger system (Schmidt et al., 2016). σ1R agonists and antagonists are classified based on their impact on σ1R’s binding affinities with its partners, such as BiP/GRP78, which often makes their classification misleading (Hayashi and Su, 2007; Kim and Bezprozvanny, 2023). The ability to potentiate neurite outgrowth in PC12 cells, and the Hill slope factor of their binding isotherms, with a slope ≈ 1 indicate an agonist. A shallow slope factor (slope ≈ 0.5) indicates an antagonist (Malar et al., 2023). Despite these methods to classify agonists and antagonist, σ1R has a single binding pocket for all ligands (agonists or antagonists; Sałaciak and Pytka, 2022), which causes the available functional assays to lack selectivity in identifying σ1R agonists or antagonists, generating further ambiguity in the classification. In conclusion, the absence of identified intrinsic activity and promiscuous binding pocket, the concept of σ1R “agonism” and “antagonism” is atypical. Therefore, the term modulator may more accurately define compounds with affinity for σ1R (Kim and Maher, 2017; Su et al., 2010). Even in the absence of a classic second messenger, the “biased signaling” may provide a framework to reconcile the seemingly contradictory effects of σ1R agonists and antagonists on MAM stability and disease outcomes. A ligand’s effect may also depend on its “functional selectivity” toward its protein partners (e.g., BiP, IP3R, GRP75 etc.) and either stabilizes or destabilizes based on the cellular context. Different ligands may also stabilize distinct σ1R conformations, each preferentially engaging in specific downstream pathways at the MAM interface. Thus, ligand-dependent conformations can yield context-specific effects rather than simple “on/off” responses. Recognizing and exploiting this biased signaling offers an opportunity to design stage-specific σ1R modulators for neurodegenerative disease intervention. Testing ligands in early vs. late disease animal models could reveal whether agonist actions help acutely and antagonist actions help chronically, or vice versa.

The lack of known intrinsic activity of σ1R suggests that targeting σ1R will have limited off-target effects. This makes σ1R ligands valuable small molecule therapeutics to treat neurological diseases including AD. σ1R ligands exert their actions through allosteric modulation of protein–protein interactions (PPIs) and signaling systems involved in multiple pathophysiological processes, including calcium dyshomeostasis, ER stress, autophagy, excitotoxicity, mitochondrial dysfunction, and reactive oxygen species (ROS) scavenging. Thus, selecting σ1R ligands targeting a specific pathology without impacting physiological functions would require a comprehensive understanding of each ligand or modulator.

Targeting the SIGMA-2 receptor (σ2R) in AD

Several anti-amnestic σ1R ligands show affinity toward its subtype sigma-2 receptor (σ2R) that has overlapping pharmacological properties. However, despite their similarities in nomenclature and ligand affinities, σ1R and σ2R are not splice variants. These receptors are products of entirely different genes on different chromosomes (Hellewell and Bowen, 1990; Izzo et al., 2014; Yi et al., 2017; Abate et al., 2015; Chu et al., 2015). More precisely, the σ1R is product of a non-opioid intracellular receptor gene located in chromosome 9. σ2R was first identified as the product of Progesterone Membrane Binding Component-1 (PGRMC1) gene, but most recent studies have concluded that σ2R gene is a product of Transmembrane 97 (TMEM97) gene (Abate et al., 2015; Chu et al., 2015). However, there is a strong possibility that both TMEM97/σ2R and PGRMC1 may be involved in the same biochemical pathways within the cell because both are implicated with cholesterol trafficking and disorder (Riad et al., 2018). Several σ1R and σ2R ligands, such as PRE-084, SA4503, and Rivastigmine, are showing promising anti-amnestic effects in pre-clinical studies of mild-to-moderate AD (Sałaciak and Pytka, 2022; Alhazmi and Albratty, 2022; Rountree et al., 2013).

We reported before that rivastigmine, a United States Food and Drug Administration (FDA)-approved anti-amnestic drug currently under preclinical studies for mild or moderate AD (Alhazmi and Albratty, 2022; Rountree et al., 2013), lowered the frequency of tight MAMs and reduced Aβ generation in vitro cellular model of AD (neuro-2A cells constitutively expressing human APP; N2AAPP) in a dose-dependent manner (Zellmer et al., 2024; Zellmer et al., 2025). This is consistent with several studies demonstrating that rivastigmine treatment lowers Aβ levels and increases neuroprotective sAPPαlevels in cultured neurons and AD mice (3XTg) (Ray et al., 2020; Bailey et al., 2011). Rivastigmine is a cholinesterase inhibitor and is found to restore neuronal plasticity in vitro via both σ1R and σ2R (Terada et al., 2018), suggesting rivastigmine as a ligand for both σ1R and σ2R. Whether rivastigmine is an agonist or antagonist for either σ1R or σ2R needs confirmation. Notably, the putative σ1R agonists are found anti-amnestic and neuroprotective, whereas the σ2R antagonists show neuroprotective property, specifically against Aβneurotoxicity. Similarly, while σ1R agonists promote anti-apoptotic signaling, σ2R antagonists block apoptosis. Therefore, despite the lack of homology, the similar binding profile and opposing activities of σ1R- and σ2R-ligands suggest that both receptors need to be considered when designing novel drugs.

σ2R is found enriched in the synapses in AD brains and hiPSC-derived neurons where σ2R colocalizes and interacts with Aβ oligomers at the synapses of AD neurons (Colom-Cadena et al., 2024). CT1812 is one of the allosteric σ2R antagonists developed by Cognitive Therapeutics Inc. (CogTx) for treating AD (Rishton et al., 2021). This drug works by displacing Aβ-oligomer from synaptic σ2R and is the first σ2R antagonist mimicking the protective effects of the Icelandic A673T mutation in the APP gene (Harper et al., 2015). CT1812 is currently in phase 1 clinical trials and is relatively safe (NCT03716427 and NCT02907567) in healthy volunteers compared to the placebo-controls (Grundman et al., 2019; Catalano et al., 2017). CT1812 has higher selectivity and affinity for the σ2R and minimal off-target effects (Izzo et al., 2014). Transcriptomic analysis revealed several differentially expressed genes (DEGs) between CT1812-treated and untreated hiPSCs+AD brain homogenates (source of Aβ oligomers; Colom-Cadena et al., 2024). Among the top-most significant transcripts, the cell adhesion Protocadherin gamma-B4 (PCDHGB4) and several glia-modulating genes and astrocytic biomarker of inflammation in AD. Pathway analysis found important role for astrocytes in protecting synapses and ultimately cognition, suggesting a role for CT1812 in modulating inflammatory pathways and restores synapse health. More detailed study of each DEGs will help uncover the mechanism of action of anti-amnestic σ2R-ligands in AD.

Although σ2R antagonists have demonstrated neuroprotective and anti-apoptotic effects, their direct role in MAM biology remains unexplored. Given the essential involvement of lipid rafts and cholesterol in MAM composition—and the known cholesterol-binding property of σ2R (Jin et al., 2022), a comprehensive investigation into the potential convergence of σ1R and σ2R in regulating MAMs could reveal synergistic mechanisms and lead to more effective therapeutic strategies for neurodegenerative diseases. Notably, Oyer et al. have provided a holistic, lipid-centric perspective on the convergence of both sigma receptors in the context of cancer treatment, offering valuable insights that may extend to neurodegenerative disease mechanisms (Oyer et al., 2023).

To date, there has been no direct evidence of whether σ2R directly influences the architecture or function of mitochondria-associated ER membranes (MAMs). One possibility is that, unlike σ1R, σ2R does not localize to or regulate MAMs. If so, this would identify a distinct downstream pathway through which σ1R and σ2R exert non-overlapping effects. Several lines of evidence suggest that despite their overlapping pharmacological profiles, σ1R and σ2R differ substantially in their biological functions (Schmidt and Kruse, 2019). For example, the two receptors have opposing roles in neuropathic pain and may similarly diverge in regulating cell survival and death (Sánchez-Blázquez et al., 2020). σ1R and σ2R also exhibit complex and partially opposing actions in adipogenesis and obesity—conditions that are rising at an alarming rate in the United States. Deletion of both receptors protects animals from diet-induced adiposity; however, the effects of single-receptor loss are sexually dimorphic. σ1R deletion protects both male and female mice, whereas σ2R deletion protects only males. Moreover, σ2R ablation but not σ₁R loss increases fatty-acid oxidation, indicating mechanistic divergence between the receptors. None of these metabolic effects are observed in female σ2R -knockout mice (Li et al., 2022), further highlighting sexual dimorphism.

Together, these findings underscore that σ1R and σ2R regulate distinct biological processes. Further mechanistic studies will be essential to determine whether their divergence extends to MAM organization or signaling, which could ultimately enable precision therapeutic strategies targeting receptor-specific pathways.

Targeting the σ1R to modulate mitochondria-associated ER membranes (MAMs) in AD

σ1R activation stabilizes MAMs and facilitates calcium transfer from the ER to mitochondria through the IP3R3-GRP75-VDAC1 channel. GRP78-free σ1R may also translocate from the MAM to other cellular compartments such as the plasma membrane, the ER membrane, and the nuclear envelope (Hayashi and Su, 2007). σ1R then interacts with various cellular interaction partners including ion channels, receptors, and kinases (Su et al., 2010). Furthermore, σ1R can translocate from MAMs to plasma membrane where it directly or indirectly modulates intracellular calcium homeostasis by regulating activities of various plasma membrane elements including N-methyl-D-aspartate receptors (NMDARs), voltage-gated calcium channels (VGCCs), acid-sensing ion channel a (ASIC1a) and stromal interaction molecules 1 (STIM1)/Orai1 complex. ER-stress or reactive oxygen species (ROS) activates σ1R resulting in its dissociation from its cognate co-chaperone BiP and stabilizes IP3R3 to form MAM-stabilizing IP3R3-GRP75-VDAC1 anchoring complex. Cancer cells express active σ1R that plays a crucial role in apoptosis by regulating [Ca2+] efflux across the ER and mitochondria after forming a complex with IP3R3 and anti-apoptotic proteins Bcl-2 and Ras-related C3 botulinum toxin substrate 1 (Rac1; Weng et al., 2017; Natsvlishvili et al., 2015). Increase Ca2+ signaling from the ER into mitochondria alters cell’s electrical plasticity, allowing the cell to become better suited for survival in a cancerous environment (Hayashi and Su, 2007).

Both MAM stabilization and calcium homeostasis are becoming increasingly relevant in AD pathogenesis. “Calcium hypothesis” is an emerging field suggesting that sustained changes in molecular mechanisms that regulate cellular [Ca2+] homeostasis, beyond the normal modulations in the cellular [Ca2+] that occur during the typical depolarization-repolarization cycles of a healthy neuron, play a critical role in age-related neurodegeneration, including AD (A.s.A.C.H. Workgroup, 2017). Polymorphism in the σ1R gene is found to be associated with ALS/FTD and AD (Uchida et al., 2005; Maruszak et al., 2007; Huang et al., 2011; Feher et al., 2012; Luty et al., 2010; Al-Saif et al., 2011; Gregianin et al., 2016; Watanabe et al., 2016). Genetic silencing of the σ1R gene in an ALS mouse model (sigmar1−/−) showed a significant reduction of MAMs in the neurons, with greater effects in axons (Bernard-Marissal et al., 2015). The putative σ1R-antagonist NE-100 dramatically reduced MAM levels, primarily in the neuronal processes or axons of a 3-dimensional human neural model of AD (Bhattacharyya et al., 2021). These studies indicate that σ1R is an upstream regulator of MAM.

As a proof-of-concept study, we reported that the highly specific σ1R agonist PRE-084 increased, while the antagonist NE-100 significantly reduced axonal Aβ release from a well-characterized 3-dimensional (3D) neural model of familial AD (FAD) (Bhattacharyya et al., 2021; Zellmer et al., 2024; Zellmer et al., 2025). Recently, we reported that the siRNA-mediated σ1R-knockdown (σ1R-KD) also reduced Aβ release from the 3D FAD model. Consistent with the results, several reports have demonstrated σ1R-antagonist actions mimicking phenotypes observed in genetic knockdown (KD) or knockout (KO) animal models (Kim and Maher, 2017; Maurice and Su, 2009; Merlos et al., 2017). The findings suggest that σ1R antagonist NE-100 may serve as small molecule therapeutics contradicting the consensus that σ1R agonists are anti-amnestic. Although, it is difficult to reconcile the discrepancies, challenges remain in identifying specific σ1R ligands due to their atypical nature lacking a second messenger system, as described above.

Direct targeting of MAM

Pharmacological inhibitors of MAM-resident SOAT1, e.g., K-604 and F12511, lowered AD pathology by increasing Aβ clearance by upregulating microglial MAMs (Shibuya et al., 2014; Shibuya et al., 2015). K-604 also reversed the microglial Aβ-clearance by reducing the levels of microglial neutral lipids in mice and hiPSCs that are deficient in AD-risk genes TREM2 or APOE (Nugent et al., 2020).

Emerging evidence suggests that in addition to regulating amyloid pathology, MAM structures also play a significant role in modulating synaptic function, which is crucial in exacerbating cognitive impairment in AD (Leung et al., 2021; Ferreira et al., 2015). MAM tethering proteins VAPB (VAMP-associated protein B) and PTPIP51(Protein Tyrosine Phosphatase Interacting Protein 51) interact at the synapses to regulate synaptic function (Gómez-Suaga et al., 2019). Defective protein synthesis in the neurons impacts synaptic plasticity, causing memory impairments in AD (Oliveira et al., 2021; Ma et al., 2013; Ribeiro et al., 2023). Counterbalancing protein synthesis in the neurons or synaptic compartments may be a potential therapeutic strategy for early treatment.

Several FDA-approved, synthetic or natural compounds showing promising results in cancer and metabolic disorders are MAM modulators (Magalhaes Rebelo et al., 2020). However, their effect on neurodegenerative diseases is not well studied. MAM modulators may be categorized into three major classes:

Class I MAM modulators

These are small-molecule reagents that target MAM structural components, e.g., gap width, length, and localization. A synthetic compound named LDC-3/Dynarrestin is one of the first-generation Class I MAM modulator that perturbs the length, gap width, and intracellular distribution of MAMs by directly targeting the mitochondrial protein tyrosine phosphatase-interacting protein 51 (PTPIP51) and disrupting its anchoring with the ER-resident vesicle-associated membrane protein-associated protein B (VAMPB; De Vos et al., 2012).

Axonal dystrophies (AxD) are key therapeutic targets that are marked by neuritic swellings and dystrophy, contributing to neurotoxic Aβ accumulation and memory loss (Leuner et al., 2012; Silva et al., 2012). A disease-related mutation (R95Q) in the MAM-tethering protein, mitofusin 2 (MFN2) disrupts mitochondrial transport and fusion, leading to the development of characteristic features of AxD while having little to no detrimental effect on the cell body (Zhou et al., 2021; Misko et al., 2012). One of the first Class I MAM modulators is a group of small-peptide mimetics, also termed mitofusin (MFN) agonists, that mimic the intramolecular interacting domains of the mitochondrial mitofusin 2 (MFN2), thereby restoring mitochondrial dynamics in neurodegenerative Charcot–Marie-Tooth type 2A disease (CMT2A) (Rocha et al., 2018). MFN2 is a well-studied MAM anchoring protein that is primarily located in the mitochondria in “closed” or “open” forms guiding mitochondrial fusion and fission, respectively. MFN2 [1–751 amino acid (aa)] forms an inactive “closed” conformation via an intramolecular interaction between the heptad repeat (HR1) domain (aa 338–418), residing between the active (GTPase) and transmembrane (TM) domains, and the C-terminal HR2 domain (aa 681–751). The “closed” conformation leads to mitochondrial fragmentation, while the “open” conformation favors mitochondrial fusion. CMT2A is the prototypical neuronal disorder of defective mitochondrial fusion and impaired mitochondrial trafficking leading to mitochondrial fragmentation and axonal degeneration. Competing peptide mimetics, e.a., Cpd A (compound A) and Cpd B (compound B), analogous to the interactive area within the MFN2 HR1 domain disrupted HR1-HR2 interaction in CMT2A neurons carrying inactive MFN2 mutations. The mimetics allosterically activated MFN2 by disrupting the HR1-HR2 interaction and converting the inactive “closed” conformation to active “open” conformation leading to the reversal of mitochondrial dysmotility, fragmentation, depolarization, and fusion. Whether the mechanism of action of these mimetics involves MAM modulation remains largely unknow.

Although MFN2 is primarily mitochondrial, approximately 5–10% is localized to the ER, where it contributes to MAM formation and function. Anchors (de Brito and Scorrano, 2008). Despite no direct association of the MFN2 gene with AD, disease-related mutation (R95Q) in MFN2 disrupted mitochondrial transport and fusion, leading to the development of characteristic features of axonal dystrophy (AxD), a key therapeutic target marked by neuritic swellings that contribute to neurotoxic Aβ accumulation and memory loss with little to no detrimental effect on the cell body (Leuner et al., 2012; Silva et al., 2012; Zhou et al., 2021; Misko et al., 2012). However, targeting MFN2 to modulate MAM formation is challenging because knocking out MFN2 in vitro or in vivo resulted in reduction of MAM formation in some studies and increase in other studies (Casellas-Díaz et al., 2021; Sugiura et al., 2013; Filadi et al., 2015; Leal et al., 2016). Therefore, whether MFN2 facilitates or impedes MAM formation remains controversial. Recent discoveries indicate that the mitochondrial MFN2 (MFN2MT) in its “open” form not only establish a homotypic contact with the 5–10% MFN2 located in the ER (MFN2ER), but also form heterotypic contacts with its isoform MFN1 found exclusively in the ER (de Brito and Scorrano, 2008). Thus, peptide mimetics targeting either the homotypic trans-dimer (MFN2MT-MFN2ER) or heterotypic trans-dimer (MFN2MT-MFN1) may have potential therapeutic properties for several neurological disorders including AD, PD, and ALS, wherein MAM structure and function are contributing factors (Bhattacharyya et al., 2021; Zellmer et al., 2024; Bernard-Marissal et al., 2015; Area-Gomez et al., 2012; Dematteis et al., 2024). Peptide mimetics are emerging as promising therapies for many neurological diseases. For example, a 24-aa peptide (p5) targeting the pathogenic Cdk5-p25 complex with remarkable specificity reduced Tau hyperphosphorylation, Aβ plaques, and improved memory and motor function in mice, without affecting the physiological Cdk5-p35 complex (Pao et al., 2023).

Class II MAM modulators

These are synthetic or natural small-molecule agents that target MAM-anchoring proteins at their transcriptional, translational, or post-translational level. Metformin is an antidiabetic drug, but long-term use is associated with a lower risk of cognitive decline among dementia patients via mechanisms unrelated to diabetes (Campbell et al., 2018). Although the results in AD are mixed, metformin reduced the risk of all types of dementia including AD if it was administered before dementia began (Ji et al., 2022). 500 mg/day administered to individuals with preclinical or mild cognitive impairment (MCI), or genetically predisposed (eg., APOEε4 homozygotes, or pathogenic SORL1) potentially modify disease progression, making it a promising candidate for repurposing in the prevention of AD. Metformin is in Alzheimer’s Dementia Prevention study (NCT04098666)—is investigating the protective effects of doses up to 2000 mg/day, with results expected in 2027. The mechanism of action of metformin is not known. However, metformin increased the expression levels of MAM-proteins VDAC1, PACS-2, and MFN2 (Guo et al., 2024; Wang et al., 2024; Foretz et al., 2014; Sanchez-Rangel and Inzucchi, 2017). Thus, metformin is a Class II MAM modulator that can be repurposed for AD (Cho et al., 2024; Daly and Imbimbo, 2025). Notably, metformin use was also associated with a significantly increased AD risk, specifically for patients with prolonged diabetes or depression (Ha et al., 2021). Therefore, a comprehensive study of metformin’s role in regulating MAM’s structural (e.g., length and gap width) and functional (e.g., Aβ-generation or clearance, lipid or calcium homeostasis, mitochondrial mobility) properties will be valuable in identifying the dose and duration of metformin treatment to prevent or lower AD risk.

In addition to metformin, several chemotherapeutics are known to either directly or indirectly modulate MAM structure/function and homeostasis. Doxorubicin, for example, is an anti-cancer drug, widely used to treat leukemias, lymphomas, and solid tumors but may develop devastating cardiomyopathy associated with reduced expression levels of the MAM anchoring protein MFN2 (Cagel et al., 2017; Tang et al., 2017). Doxorubicin can also decrease the expression of the anti-apoptotic Bcl 2, enhancing cell death stimuli in breast cancer cell lines (Pilco-Ferreto and Calaf, 2016).

Cisplatin (CIS) is a chemotherapeutic agent potentially functioning as a MAM modulator because its anti-cancer effect is mediated by IP3R (Tsunoda et al., 2005). Moreover, CIS is in a complex feedback mechanism involving the downregulation of MFN2 and GRP75 (Xie et al., 2016). Thus, CIS may be repurposed to treat neurological diseases with MAM pathology. However, the biggest obstacle in repurposing is that the anti-cancer dose of CIS develops CIS resistance and cause serious damage to the brain (Cicek et al., 2024). A comprehensive study of CIS’s impact on MAM architecture (“tightening” or “loosening” effects) may uncover avenues to repurpose this drug without the major side effects.

A recent study has identified the antiepileptic drug clemastine as a potential candidate for repurposing in other neurological disorders (Badawi et al., 2024), including AD, given that epilepsy pathophysiology is strongly influenced by elevated Aβ expression levels (Hickman et al., 2023). Clemastine has previously been shown to prevent cognitive dysfunction in preclinical models of multiple sclerosis and cerebral ischemia (Sałaciak and Pytka, 2022). Mechanistically, clemastine helps maintain cellular homeostasis and prevent cell death through activation of σ1R. Activation of σ1R influences multiple pathological pathways, including calcium homeostasis, ER stress, autophagy, excitotoxicity, mitochondrial dysfunction, and reactive oxygen species (ROS) clearance, thereby exerting broad neuroprotective effects against neuroinflammation, neuronal excitotoxicity and apoptosis (Ruiz-Cantero et al., 2021).

Class III MAM modulators

These targets upstream regulator(s) of MAM stabilization. σ1R is one of the first upstream regulators of MAM’s structure and function. As described above, numerous studies have established that the σ1R is one of the major therapeutic targets against AD (Piechal et al., 2021), and found that σ1R regulates amyloid pathology by modulating MAMs’ plasticity, specifically their lengths, gap widths, somal vs. axonal distribution, and axonal mobility (Bhattacharyya et al., 2021; Zellmer et al., 2024; Zellmer et al., 2025). The impacts of the putative σ1R-antagonist NE-100 and σ1R/σ2R-ligand rivastigmine on Aβ levels in a 3D neural model of AD describes a novel σ1R/σ2R-MAM axis in regulating amyloid pathology in AD (Bhattacharyya et al., 2021; Zellmer et al., 2024). The innovative method of measuring mitochondrial axonal velocity as a useful mean to quantify MAM gap width stabilization has provided high precision in identifying modulators of σ1R/σ2R-MAM axis to delay or prevent AD pathogenesis. Numerous pharmaceutical drugs and small molecules exhibiting diverse chemical structures and therapeutic/pharmacological profiles act as σ1R or σ2R ligands (Weber and Wünsch, 2017; Fallica et al., 2021). Many σ1R/σ2R ligands (agonists/antagonists/modulators) have been FDA-approved, which could be repurposed for the treatment of AD and ADRDs (Table 1).

Table 1
www.frontiersin.org

Table 1. Small-molecule σ1R/σ2R modulators: investigational therapeutics for AD and AD-related disorders (ADRD).

MAM gap width is a potential therapeutic target

The architecture of MAMs—particularly the gap width between the ER and mitochondria, which ranges from 6 to 80 nm—plays a crucial role in regulating their function (Giacomello and Pellegrini, 2016; Sukhorukov et al., 2022; Zhang et al., 2021; Cieri et al., 2018; Carpio et al., 2021; Csordas et al., 2006; Ziegler et al., 2021). Transmission electron microscopy (TEM) studies reveal that smooth ER (SER) typically forms “tight” MAMs with 8–15 nm gaps, while rough ER (RER) forms wider contacts of 20–30 nm; in some contexts, the gap can reach up to 80 nm (Filadi et al., 2015; Hirabayashi et al., 2017; Kowaltowski et al., 2019). Notably, axonal ER is predominantly SER, and therefore primarily forms tight MAMs (Öztürk et al., 2020).

MAM gap width defines MAM structure and function. Tight MAMs (~10 nm) are pro-apoptotic and amyloidogenic, whereas medium (~25 nm) or loose (25–80 nm) MAMs appear anti-apoptotic and anti-amyloidogenic (Zellmer et al., 2024; Prudent et al., 2015). In AD, both the structure and function of MAMs, such as their horizontal length or vertical gap width and Aβ generation are perturbed in AD (Figure 1; Schon and Area-Gomez, 2013; Erpapazoglou et al., 2017; Leal et al., 2020). Increased MAM formation has been detected in fibroblasts and post-mortem brain tissue from familial and sporadic AD patients, and AD-associated proteins (APP, BACE1, and γ-secretase components) are enriched at these sites patients (Pera et al., 2017; Area-Gomez et al., 2009; Area-Gomez et al., 2012). Our previous work showed that σ1R knockdown reduced tight MAMs (<10 nm) and increased loose MAMs (~2.5-fold), mirroring findings in AD transgenic neurons (Zellmer et al., 2024; Martino Adami et al., 2019). Using optic nerves from mouse and human models—purely axonal systems—we found a > 4-fold increase in tight MAMs in AD axons relative to controls (Zellmer et al., 2024). Using mouse and human optic nerves, reliable in vivo models of pure axons, we observed a > 4-fold increase in tight MAMs in AD axons compared to controls (Zellmer et al., 2024). These findings highlight MAM architecture as a potential therapeutic target (Figure 2).

Figure 1
Electron micrographs depict mitochondria-associated membranes (MAMs) with different gap widths: less than 10 nanometers highlighted in magenta (tight MAMs), 10 to 80 nanometers in yellow (loose MAMs), and no MAM at more than 80 nanometers. A bar chart shows percentages of MAMs per mitochondrion for different gap widths (tight and loose), compared between si-non and si-s1R groups, with si-s1R showing lower percentages of tight and higher percentages of loose MAMs.

Figure 1. σ1R knockdown decreased the frequency of tight MAMs and lowered Aβ generation in vitro. (A) TEM images showing ER (ER)-mitochondria (M) contact sites or MAMs of gap widths < 10 nm (purple) or > 10 nm (orange) in ReN-GA cells electroporated with scrambled siRNA (si-non) or siRNA against σ1R (si-σ1R) for 48 h. Mitochondria separated by > 80 nm from the ER are non-MAMs. (B) Quantitation of the tight (< 10 nm) or loose (> 10 nm) MAMs in control (si-non) and σ1R-silenced (si-σ1R) cells per mitochondria per frame (MAM per mitochondria). More than seven frames were used for each analysis. An average of 3–6 mitochondria forming tight or loose MAMs from each frame were manually counted (unbiased, *p < 0.05; **p < 0.001; Bhattacharyya et al., 2021; Zellmer et al., 2024).

Figure 2
Illustration showing three conditions of mitochondria associated membranes (MAM). From left to right: no-MAM state with over eighty nanometer distance, basal/loose MAM with twenty to forty nanometer distance, and pathological state with ten nanometer distance. Proteins labeled include VDAC1, Grp75, and others. Arrows indicate transitions and effects of σ1R modulators.

Figure 2. Schematic of σ1R modulators “loosening” the pathogenic tight MAMs to basal or therapeutic loose MAMs. The mitochondria and ER form MAMs (~20 nm gap width) via σ1R-mediated formation of the IP3R3-Grp75-VDAC1 anchor. Abnormal or un-inhibited σ1R results in “tightening” the MAM gap width (<10 nm) and stabilizing tight MAMs that are pathogenic (Aβ-increasing). σ1R modulators reverses the pathogenic tight MAMs to basal or therapeutic MAMs of gap widths ranging from 20 to 40 nm.

Pharmacological and natural compounds can “tighten” or “loosen” MAMs, and several are currently in preclinical or clinical trials for metabolic disorders and cancer (Magalhaes Rebelo et al., 2020). We showed that disrupting MAM gap width by knocking down the σ1R, introducing constitutive linkers designed to stabilize MAMs of different gap widths, or inducible MAM-tightening biosensors exacerbated Aβ accumulation in a human stem cell-derived neural progenitor ReN cell model overexpressing familial AD-mutant of Aβ-precursor protein, APPSwe/Lon (ReN GA cell-derived neural culture in 3D; Bhattacharyya et al., 2021; Zellmer et al., 2024). Moreover, tightly fused ER–mitochondrial contacts significantly (p < 0.001) impaired both anterograde and retrograde axonal transport of mitochondria, whereas medium MAMs (~25 nm) maintained normal dynamics (Table 2; Zellmer et al., 2024; Zellmer et al., 2025). This observation aligns with reduced mitochondrial trafficking in AD cortical neurons (Dai et al., 2002) and AD cybrid cells (Trimmer and Borland, 2005). Thus, promoting “loose” MAMs could restore mitochondrial motility and shift pathogenic Aβ-producing MAMs toward a more homeostatic state.

Table 2
www.frontiersin.org

Table 2. Percent axonal movement (overall, retrograde, anterograde).

One of the most innovative aspects of our finding is the development of a novel and reliable system to quantify the degree of stabilization of MAMs of different gap widths, circumventing the limitations of traditional techniques like the transmission electron microscopy (TEM), cryo-TEM, or Scanning Electron Microscopies (SEM). TEM and SEM may detect cellular structures at the nanoscale level but cannot quantify MAM’s the degree of MAM stabilization due to the dynamic nature of MAMs (Cieri et al., 2018; Giacomello and Pellegrini, 2016; Stacchiotti et al., 2018). Thus, we have developed an innovative live-cell system that quantifies MAM stabilization by using mitochondrial axonal velocity as a proxy for MAM gap width. Our approach provides a functional metric to screen for therapeutic σ1R ligands for AD. For instance, σ1R ligands that yield average mitochondrial speeds of 0.6–0.7 μm/s (vs. < 0.3 μm/s) may act as small-molecule therapeutics to lower amyloid pathology.

MAM gap width also plays a significant role in modulating mitochondrial calcium levels [Ca2+MT] and regulating the programmed cell death by modulating IP3R3 forming a complex with the pro-apoptotic BOK and mitochondrial VDAC1 at the MAM. Tight MAMs, termed “full MAMs” are pro-apoptotic, while loose MAMs, denoted as “defective MAMs” reverse apoptosis (Prudent et al., 2015).

Thus, while a σ1R-ligand exhibiting average mitochondrial axonal speed ~0.7 μm/s may serve as anti-amyloid therapeutic (Table 2; Zellmer et al., 2024), we predict that σ1R-ligands exhibiting average mitochondrial axonal speed ~0.3 μm/s will serve as anti-cancer drug. However, we must highlight that the MAM gap width does not always follow a strictly linear relationship with MAM function. While narrowing the gap from >35 nm to ~20 nm enhanced [Ca2+MT] levels, further tightening below 10 nm reversed the effect (Katona et al., 2022). Therefore, identifying an optimal “therapeutic gap width” may be key to modulating ER–mitochondrial communication safely. Precisely tuning MAM architecture through σ1R ligands could enable targeted interventions to prevent or delay the onset of neurodegenerative or metabolic diseases. Targeting σ1R, an upstream modulator of MAM is safer and efficacious compared to directly targeting MAMs because of their structural and functional diversities. However, the emerging field of structural systems pharmacology (Duran-Frigola et al., 2013; Berger and Iyengar, 2011), which considers the specific properties of the drug targets and their environment, offers promise in developing effective MAM modulators for therapeutic purposes in the future.

A key contributor to AD pathology is the disruption of intracellular calcium (Ca2+) homeostasis (Green, 2008). Aβ increases cytosolic Ca2+ ([Ca2+CT]) levels, leading to elevated mitochondrial Ca2+ ([Ca2+MT]), which further promotes Aβ accumulation and neuronal death (Calvo-Rodriguez et al., 2020). Under physiological conditions, the ER maintains Ca2+ balance through the sarco/ER Ca2+ ATPase isoform SERCA2b, which pumps Ca2+ from the cytosol into the ER (Figure 3A) (Britzolaki et al., 2018).

Figure 3
Diagram illustrating mitochondria-associated membranes (MAM) and calcium signaling between mitochondria and the endoplasmic reticulum. Panel A shows the mitochondrion with outer and inner mitochondrial membranes. Panel B explains the calcium hypothesis with calcium ion transfer. Panel C relates to cancer and neurodegeneration, showing proteins like IP3R3, σ1R, Bcl-2, and BOK affecting apoptosis. Panel D presents the MAM hypothesis, highlighting the roles of proteins like APP, Aβ, BACE1, and γ-secretases in processing. The diagram highlights cellular pathways and protein interactions.

Figure 3. Therapeutic σ1R ligand for amyloidosis or oncogenesis. (A) The ER-resident SERCA regulates ER → cytoplasmic calcium transport via Calnexin and TMX1. (B) In physiological condition the σ1R remains bound to BiP/GRP78. Upon stimulation (ER- or oxidative-stress) σ1R dissociates from BiP/GRP78 and forms IP3R3-GRP75-VDAC1 anchoring complex to stabilize. VDAC1 then interacts with mitochondrial calcium uniporter (MCU) and promotes ER → mitochondria Ca2+-efflux. The “Calcium Hypothesis” of AD asserts that sustained changes in molecular mechanisms that regulate Ca2+ homeostasis, play a critical role in several chronic age-related brain disorders, such as AD. (C) MAM-tightening/loosening results in increased IP3R3 interaction either with the pro-apoptotic BOK or anti-apoptotic Bcl-2. Inhibiting or enhancing σ1R-stimulation by σ1R-ligands will impact apoptosis, a key molecular pathway that regulates oncogenesis (cancer) or neuronal death (neurodegeneration). (D) APP, BACE1, γ-secretases as well as C99 and Aβ are all found in MAM fractions. Would increase BACE1-mediated amyloidogenic processing of APP. In contrast, σ1R ligands that maintain “looser” MAM contacts may exert anti-amyloidogenic effects. This aligns with the MAM hypothesis, which posits that MAM perturbation is an early pathological event that remains persistent throughout the disease progression.

MAM gap width critically regulates Ca2+ transport. When MAMs are loose (>35 nm), Ca2+ transfer from ER to mitochondria is impaired. Tightening the MAMs to their normal gap width (~20–25 nm) restores physiological Ca2+ flux, while excessive tightening (<10 nm) severely reduces mitochondrial Ca2+ uptake (Carpio et al., 2021; Katona et al., 2022). Thus, MAM stability modulates ER–mitochondria Ca2+ exchange ([Ca2+ER] [Ca2+MT]) in a non-linear manner. Interestingly, modifying MAMs, either by tightening or loosening the gap widths does not alter cytosolic [Ca2+CT]. However, the mechanisms driving accelerated mitochondrial Ca2+ uptake under pathological conditions remain unclear. Under physiological conditions, the σ1R is bound to BiP/GRP78 in the ER. During ER or oxidative stress, σ1R dissociates and forms an IP3R3–GRP75–VDAC1 complex that tightens MAMs and stabilizes ER–mitochondria communication. VDAC1 then interacts with the mitochondrial calcium uniporter (MCU) to promote Ca2+ transfer from the ER to mitochondria ([Ca2+ER] → [Ca2+MT]; Figure 3B).

Supporting our “Therapeutic Gap Width” hypothesis, recent evidence in human iPSC-derived astrocytes from PD patients shows that stabilizing the MAM gap width at 20 nm, but not at 10 nm, fully restores mitochondrial Ca2+ uptake (Dematteis et al., 2024). This finding directly demonstrates that ER-mitochondria gap width is a critical and tunable determinant of mitochondrial Ca2+ uptake and metabolism. This finding is consistent with our proposed model, “tightening” the MAM gap below 20 nm is pathogenic (Aβ-increasing), whereas “loosening” it beyond 25 nm may be therapeutic (Aβ-lowering) (Zellmer et al., 2024; Zellmer et al., 2025). Therefore, normalizing ER-mitochondrial Ca2+ transfer or reducing Aβ generation by modulating the MAM gap width may offer a universal therapeutic strategy to restore cellular homeostasis across disorders marked by impaired mitochondrial function or mobility. These findings introduce a new paradigm in which fine-tuning the ER–mitochondria apposition to an optimal gap width could reverse mitochondrial dysfunction, such as dysmotility, fragmentation, depolarization, and defective fusion, implicated in AD (D’Alessandro et al., 2025).

MAM stabilization also influences apoptosis by regulating interactions between IP3R3 and the pro-apoptotic protein BOK or the anti-apoptotic protein Bcl-2 (Figure 3C, upper panel; Carpio et al., 2021). Through these interactions, σ1R-mediated MAM modulation controls programmed cell death—a key pathway linked to oncogenesis.

The major amyloidogenic components, APP, BACE1, γ-secretases as well as the BACE1-cleaved neurotoxic C-terminal 99 amino acid fragment of APP (C99) and γ-secretase-cleaved Aβ are all found in the purified MAM fractions in vitro and in vivo (Pera et al., 2017; Area-Gomez et al., 2009; Area-Gomez and Schon, 2017). MAM-tightening increased both BACE1-mediated APP cleavage and Aβ generation (Figure 3D, lower panel; Bhattacharyya et al., 2021; Zellmer et al., 2024). Therefore, σ1R ligands that stabilize MAMs at the optimal gap width (20–25 nm) may help maintain physiological Ca2+ homeostasis by balancing pro- and anti-apoptotic signaling. Such ligands could have potential therapeutic value as anti-cancer agents by promoting controlled, pro-apoptotic MAM stabilization.

Future direction

Future research should aim to elucidate the pathway linking MAM modulation to learning and different stages of memory. To date, no studies have directly examined how MAM plasticity (analogous to mitochondrial plasticity (Comyn et al., 2024) in its ability to undergo dynamic changes in gap width, thickness, length, and function) contributes to cellular and network-level neural substrates of learning and memory through σ1R modulation.

We argue that MAMs are critical for regulating the cellular and molecular substrates of brain plasticity underlying various forms of learning, as well as spatial and non-spatial memory. This argument is supported by well-established in vitro and in vivo evidence showing that modulation of σ1R and mitochondrial plasticity influences memory-related plasticity mechanisms like hippocampal neurogenesis, neuronal spine remodeling, and long-term potentiation and depression (Comyn et al., 2024; Maurice and Goguadze, 2017). Moreover, σ1R interacts with plasticity-related proteins such as cAMP response element-binding protein, c-fos, kinases, and brain-derived neurotrophic factor (BDNF), and modulate both cholinergic and glutamatergic neurotransmission which contributes to memory encoding, consolidation, and retrieval (Maurice and Goguadze, 2017; Crouzier et al., 2020). However, whether this learning-related and learning-induced plasticity is mediated through MAMs remains unknown.

Furthermore, it would be valuable to identify the pathway linking MAM modulation to memory through system-level investigations by integrating in vivo electrophysiology and neuroimaging approaches under σ1R modulation and how it relates to cellular plasticity mechanisms. For example, in rodent models, assessing network level plasticity during and after learning by recording regional and simultaneous interregional electrophysiological dynamics (like local field potentials, coherence, and connectivity) within early- AD affected regions like entorhinal cortex, hippocampus and their communication with prefrontal cortex, under regionally localized or systemic σ1R modulation could reveal role of MAM plasticity in behavior. Region-specific delivery of σ1R modulators targeting hippocampal circuits, followed by assessment of electrophysiological correlates such as theta oscillatory power, theta–gamma coupling, and hippocampal–frontal connectivity would be worthwhile.

Furthermore, PET imaging of σ1R in live mouse brains using the [11C]CNY-01 probe, as described above, can help identify and quantify the spatial distribution of MAMs across different brain regions under σ1R modulation. Mapping region-specific alterations in pathological MAMs may offer critical insights into the early mechanisms driving AD pathology and help determine whether targeted delivery of σ1R modulators is warranted.

Finally, an investigation into how the environmental and lifestyle factors implicated in AD - such as sleep deprivation, smoking, particulate matter exposure, physical inactivity etc. affect MAM plasticity and whether these changes mediate cognitive deficits in AD could provide valuable insights for preclinical modeling and therapeutic development targeting MAMs. Testing a hypothesis that fear-conditioning learning induces a transient loosening of MAMs in the hippocampus, a process that is disrupted in AD models, can be restored through σ1R modulation. The results of this investigation may connect the established effects of σ1R on mitochondrial function and ER stress, thereby offering a mechanistic link to MAM plasticity.

Conclusion

The three FDA-approved anti-Aβ immunotherapies (Lequembi, Aduhelm, and Kisunla) lowered AD risks of patients with prodromal or mild AD underscoring the clinical importance of targeting Aβ before symptoms appear (Malar et al., 2023). Additionally, anti-Aβ immunotherapies, e.g., Gamunex, Remternetug, and Sabitnetug have reached Phase 2/3 clinical trials for patients with early AD. The modest efficacy of anti-Aβ immunotherapies in early-stage AD emphasizes that while Aβ remains a relevant target, the real opportunity lies in intervening at the earliest disease stages. The findings are consistent with several preclinical and clinical studies indicating that the therapeutic intervention before symptoms is a more promising approach to prevent or delay AD pathology than targeting after symptoms appear (Hampel et al., 2021; Granzotto and Sensi, 2024). Despite showing promising results, the high cost and significant safety concerns of anti-Aβ immunotherapies highlight the need for safer, cost-effective, and mechanistically distinct alternative therapeutic strategies (van Dyck et al., 2023; Budd Haeberlein et al., 2022).

Synthetic or natural small molecule ligands of σ1R and σ2R are showing promising results in treating neurological disorders (Malar et al., 2023). Given the architectural diversity of MAMs and their critical role in AD pathology, targeting σ1R, an upstream modulator of MAM’s structure offers a novel and potentially safer alternative to traditional anti-Aβ therapies. σ1R appears to be one of the most significant and underexplored therapeutic targets in AD, specifically at the early stages of AD. σ1R’s therapeutic action may involve complex, context-dependent mechanisms targeting either extracellular or axonal Aβ to treat AD at the early stages. The lack of understanding of σ1R-mediated dynamic regulation of MAMs during AD progression and the mechanistic basis for its modulatory effects on Aβ metabolism are critical barriers to therapeutic optimization. Dissecting the intersection of σ1R signaling, ER-mitochondrial communication, and Aβ pathology in AD brains by studying the impacts of σ1R modulation on MAM structure and function will not only advance AD treatment but may also reveal a unifying framework for targeting shared pathogenic pathways across multiple neurological disorders linked to ER-mitochondrial dysfunction.

The focus of this mini review is to examine the convergence between sigma receptors, particularly σ1R, and MAM structure–function as a mechanism underlying disease modification in AD, ALS, PD, and HD. Although interest in the role of sigma receptors in other neurodegenerative diseases is growing, a direct link between sigma receptor activity and MAM structural or functional dynamics has not yet been established. Two recent reviews have highlighted the pleiotropic mechanism of actions of σ1R/σ2R ligands, including modulation of cellular stress and excitotoxicity, which have shown benefits in conditions such as stroke, epilepsy, neuropathic pain, and psychiatric disorders (Piechal et al., 2021; Drewes et al., 2025; Ruiz-Cantero et al., 2024). Together, these findings reinforce the concept that σ1R/σ2R act as master regulators of cellular resilience and represent promising therapeutic targets across a wide spectrum of CNS diseases.

Author contributions

ML: Writing – review & editing, Data curation. JZ: Writing – review & editing, Data curation, Methodology. RB: Conceptualization, Writing – review & editing, Methodology, Supervision, Data curation, Writing – original draft, Investigation, Software, Project administration, Funding acquisition, Visualization, Resources, Formal analysis, Validation.

Funding

The author(s) declared that financial support was received for this work and/or its publication. Cure Alzheimer’s Fund (to RB) and funding number is GR0244415.

Acknowledgements

We thank the Cure Alzheimer’s Fund (CAF) grant (to RB) for the support.

Conflict of interest

The author(s) declared that this work was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declared that Generative AI was used in the creation of this manuscript. ChatGPT was used to shorten the abstract.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

A.S.A.C.H. Workgroup (2017). Calcium hypothesis of Alzheimer's disease and brain aging: a framework for integrating new evidence into a comprehensive theory of pathogenesis. Alzheimers Dement. 13, 178–182.e17. doi: 10.1016/j.jalz.2016.12.006,

PubMed Abstract | Crossref Full Text | Google Scholar

Abate, C., Niso, M., Infantino, V., Menga, A., and Berardi, F. (2015). Elements in support of the ‘non-identity’ of the PGRMC1 protein with the σ2 receptor. Eur. J. Pharmacol. 758, 16–23. doi: 10.1016/j.ejphar.2015.03.067,

PubMed Abstract | Crossref Full Text | Google Scholar

Alhazmi, H. A., and Albratty, M. (2022). An update on the novel and approved drugs for Alzheimer disease. Saudi Pharm J 30, 1755–1764. doi: 10.1016/j.jsps.2022.10.004,

PubMed Abstract | Crossref Full Text | Google Scholar

Al-Saif, A., Al-Mohanna, F., and Bohlega, S. (2011). A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann. Neurol. 70, 913–919. doi: 10.1002/ana.22534,

PubMed Abstract | Crossref Full Text | Google Scholar

Area-Gomez, E., de Groof, A. J., Boldogh, I., Bird, T. D., Gibson, G. E., Koehler, C. M., et al. (2009). Presenilins are enriched in endoplasmic reticulum membranes associated with mitochondria. Am. J. Pathol. 175, 1810–1816. doi: 10.2353/ajpath.2009.090219,

PubMed Abstract | Crossref Full Text | Google Scholar

Area-Gomez, E., Del Carmen Lara Castillo, M., Tambini, M. D., Guardia-Laguarta, C., de Groof, A. J., Madra, M., et al. (2012). Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. EMBO J. 31, 4106–4123. doi: 10.1038/emboj.2012.202,

PubMed Abstract | Crossref Full Text | Google Scholar

Area-Gomez, E., and Schon, E. A. (2017). On the pathogenesis of Alzheimer's disease: the MAM hypothesis. FASEB J. 31, 864–867. doi: 10.1096/fj.201601309,

PubMed Abstract | Crossref Full Text | Google Scholar

Badawi, G. A., Shokr, M. M., Elshazly, S. M., Zaki, H. F., and Mohamed, A. F. (2024). Sigma-1 receptor modulation by clemastine highlights its repurposing as neuroprotective agent against seizures and cognitive deficits in PTZ-kindled rats. Eur. J. Pharmacol. 980:176851. doi: 10.1016/j.ejphar.2024.176851,

PubMed Abstract | Crossref Full Text | Google Scholar

Bai, P., Gomm, A., Yoo, C.-H., Mondal, P., Lobo, F. M., Meng, H., et al. (2025). Development of carbon-11 labeled pyrimidine derivatives as novel positron emission tomography (PET) agents enabling brain sigma-1 receptor imaging. Adv. Sci. 12:2414827. doi: 10.1002/advs.202414827

Crossref Full Text | Google Scholar

Bailey, J. A., Ray, B., Greig, N. H., and Lahiri, D. K. (2011). Rivastigmine lowers aβ and increases sAPPα levels, which parallel elevated synaptic markers and metabolic activity in degenerating primary rat neurons. PLoS One 6:e21954. doi: 10.1371/journal.pone.0021954,

PubMed Abstract | Crossref Full Text | Google Scholar

Berger, S. I., and Iyengar, R. (2011). Role of systems pharmacology in understanding drug adverse events. Wiley Interdiscip. Rev. Syst. Biol. Med. 3, 129–135. doi: 10.1002/wsbm.114,

PubMed Abstract | Crossref Full Text | Google Scholar

Bernard-Marissal, N., Medard, J. J., Azzedine, H., and Chrast, R. (2015). Dysfunction in endoplasmic reticulum-mitochondria crosstalk underlies SIGMAR1 loss of function mediated motor neuron degeneration. Brain 138, 875–890. doi: 10.1093/brain/awv008,

PubMed Abstract | Crossref Full Text | Google Scholar

Bhattacharyya, R., Barren, C., and Kovacs, D. M. (2013). Palmitoylation of amyloid precursor protein regulates amyloidogenic processing in lipid rafts. J. Neurosci. 33, 11169–11183. doi: 10.1523/JNEUROSCI.4704-12.2013,

PubMed Abstract | Crossref Full Text | Google Scholar

Bhattacharyya, R., Black, S. E., Lotlikar, M. S., Fenn, R. H., Jorfi, M., Kovacs, D. M., et al. (2021). Axonal generation of amyloid-beta from palmitoylated APP in mitochondria-associated endoplasmic reticulum membranes. Cell Rep. 35:109134. doi: 10.1016/j.celrep.2021.109134,

PubMed Abstract | Crossref Full Text | Google Scholar

Britzolaki, A., Saurine, J., Flaherty, E., Thelen, C., and Pitychoutis, P. M. (2018). The SERCA2: A gatekeeper of neuronal calcium homeostasis in the brain. Cell. Mol. Neurobiol. 38, 981–994. doi: 10.1007/s10571-018-0583-8,

PubMed Abstract | Crossref Full Text | Google Scholar

Budd Haeberlein, S., Aisen, P. S., Barkhof, F., Chalkias, S., Chen, T., Cohen, S., et al. (2022). Two randomized phase 3 studies of aducanumab in early Alzheimer's disease. J. Prev Alzheimers Dis. 9, 197–210. doi: 10.14283/jpad.2022.30,

PubMed Abstract | Crossref Full Text | Google Scholar

Cagel, M., Grotz, E., Bernabeu, E., Moretton, M. A., and Chiappetta, D. A. (2017). Doxorubicin: nanotechnological overviews from bench to bedside. Drug Discov. Today 22, 270–281. doi: 10.1016/j.drudis.2016.11.005,

PubMed Abstract | Crossref Full Text | Google Scholar

Calvo-Rodriguez, M., Hou, S. S., Snyder, A. C., Kharitonova, E. K., Russ, A. N., Das, S., et al. (2020). Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer's disease. Nat. Commun. 11:2146. doi: 10.1038/s41467-020-16074-2,

PubMed Abstract | Crossref Full Text | Google Scholar

Campbell, J. M., Stephenson, M. D., de Courten, B., Chapman, I., Bellman, S. M., and Aromataris, E. (2018). Metformin use associated with reduced risk of dementia in patients with diabetes: A systematic review and Meta-analysis. J Alzheimer's Dis 65, 1225–1236. doi: 10.3233/JAD-180263,

PubMed Abstract | Crossref Full Text | Google Scholar

Carpio, M. A., Means, R. E., Brill, A. L., Sainz, A., Ehrlich, B. E., and Katz, S. G. (2021). BOK controls apoptosis by ca(2+) transfer through ER-mitochondrial contact sites. Cell Rep. 34:108827. doi: 10.1016/j.celrep.2021.108827,

PubMed Abstract | Crossref Full Text | Google Scholar

Casellas-Díaz, S., Larramona-Arcas, R., Riqué-Pujol, G., Tena-Morraja, P., Müller-Sánchez, C., Segarra-Mondejar, M., et al. (2021). Mfn2 localization in the ER is necessary for its bioenergetic function and neuritic development. EMBO Rep. 22:e51954. doi: 10.15252/embr.202051954,

PubMed Abstract | Crossref Full Text | Google Scholar

Catalano, S., Grundman, M., Schneider, L. S., DeKosky, S., Morgan, R., Guttendorf, R., et al. (2017). [P4–567]: A phase 1 safety trial of the aβ oligomer receptor antagonist CT1812. Alzheimers Dement. 13:P1570. doi: 10.1016/j.jalz.2017.07.730

Crossref Full Text | Google Scholar

Cho, S. Y., Kim, E. W., Park, S. J., Phillips, B. U., Jeong, J., Kim, H., et al. (2024). Reconsidering repurposing: long-term metformin treatment impairs cognition in Alzheimer’s model mice. Transl. Psychiatry 14:34. doi: 10.1038/s41398-024-02755-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Chu, U. B., Mavlyutov, T. A., Chu, M.-L., Yang, H., Schulman, A., Mesangeau, C., et al. (2015). The Sigma-2 receptor and progesterone receptor membrane component 1 are different binding sites derived from independent genes. EBioMedicine 2, 1806–1813. doi: 10.1016/j.ebiom.2015.10.017,

PubMed Abstract | Crossref Full Text | Google Scholar

Cicek, B., Danisman, B., Bolat, I., Kiliclioglu, M., Kuzucu, M., Suleyman, H., et al. (2024). Effect of tangeretin on cisplatin-induced oxido-inflammatory brain damage in rats. J. Cell. Mol. Med. 28:e18565. doi: 10.1111/jcmm.18565,

PubMed Abstract | Crossref Full Text | Google Scholar

Cieri, D., Vicario, M., Giacomello, M., Vallese, F., Filadi, R., Wagner, T., et al. (2018). SPLICS: a split green fluorescent protein-based contact site sensor for narrow and wide heterotypic organelle juxtaposition. Cell Death Differ. 25, 1131–1145. doi: 10.1038/s41418-017-0033-z,

PubMed Abstract | Crossref Full Text | Google Scholar

Colom-Cadena, M., Toombs, J., Simzer, E., Holt, K., McGeachan, R., Tulloch, J., et al. (2024). Transmembrane protein 97 is a potential synaptic amyloid beta receptor in human Alzheimer’s disease. Acta Neuropathol. 147:32. doi: 10.1007/s00401-023-02679-6,

PubMed Abstract | Crossref Full Text | Google Scholar

Comyn, T., Preat, T., Pavlowsky, A., and Placais, P. Y. (2024). Mitochondrial plasticity: an emergent concept in neuronal plasticity and memory. Neurobiol. Dis. 203:106740. doi: 10.1016/j.nbd.2024.106740,

PubMed Abstract | Crossref Full Text | Google Scholar

Crouzier, L., Couly, S., Roques, C., Peter, C., Belkhiter, R., Arguel Jacquemin, M., et al. (2020). Sigma-1 (sigma(1)) receptor activity is necessary for physiological brain plasticity in mice. Eur. Neuropsychopharmacol. 39, 29–45. doi: 10.1016/j.euroneuro.2020.08.010

Crossref Full Text | Google Scholar

Csordas, G., Renken, C., Varnai, P., Walter, L., Weaver, D., Buttle, K. F., et al. (2006). Structural and functional features and significance of the physical linkage between ER and mitochondria. J. Cell Biol. 174, 915–921. doi: 10.1083/jcb.200604016,

PubMed Abstract | Crossref Full Text | Google Scholar

D’Alessandro, M. C. B., Kanaan, S., Geller, M., Praticò, D., and Daher, J. P. L. (2025). Mitochondrial dysfunction in Alzheimer’s disease. Ageing Res. Rev. 107:102713. doi: 10.1016/j.arr.2025.102713,

PubMed Abstract | Crossref Full Text | Google Scholar

Dai, J., Buijs, R. M., Kamphorst, W., and Swaab, D. F. (2002). Impaired axonal transport of cortical neurons in Alzheimer’s disease is associated with neuropathological changes. Brain Res. 948, 138–144. doi: 10.1016/S0006-8993(02)03152-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Daly, T., and Imbimbo, B. P. (2025). Enhancing uptake of antidiabetic drugs to reduce cardiometabolic risk and dementia burden. Alzheimers Dement. 21:e70614. doi: 10.1002/alz.70614,

PubMed Abstract | Crossref Full Text | Google Scholar

de Brito, O. M., and Scorrano, L. (2008). Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456, 605–610. doi: 10.1038/nature07534,

PubMed Abstract | Crossref Full Text | Google Scholar

De Vos, K. J., Morotz, G. M., Stoica, R., Tudor, E. L., Lau, K. F., Ackerley, S., et al. (2012). VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum. Mol. Genet. 21, 1299–1311. doi: 10.1093/hmg/ddr559,

PubMed Abstract | Crossref Full Text | Google Scholar

Dematteis, G., Tapella, L., Casali, C., Talmon, M., Tonelli, E., Reano, S., et al. (2024). ER-mitochondria distance is a critical parameter for efficient mitochondrial Ca2+ uptake and oxidative metabolism. Commun. Biol. 7:1294. doi: 10.1038/s42003-024-06933-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Drewes, N., Fang, X., Gupta, N., and Nie, D. (2025). Pharmacological and pathological implications of Sigma-1 receptor in neurodegenerative diseases. Biomedicine 13:1409. doi: 10.3390/biomedicines13061409,

PubMed Abstract | Crossref Full Text | Google Scholar

Duran-Frigola, M., Mosca, R., and Aloy, P. (2013). Structural systems pharmacology: the role of 3D structures in next-generation drug development. Chem. Biol. 20, 674–684. doi: 10.1016/j.chembiol.2013.03.004,

PubMed Abstract | Crossref Full Text | Google Scholar

Erpapazoglou, Z., Mouton-Liger, F., and Corti, O. (2017). From dysfunctional endoplasmic reticulum-mitochondria coupling to neurodegeneration. Neurochem. Int. 109, 171–183. doi: 10.1016/j.neuint.2017.03.021,

PubMed Abstract | Crossref Full Text | Google Scholar

Fallica, A. N., Pittalà, V., Modica, M. N., Salerno, L., Romeo, G., Marrazzo, A., et al. (2021). Recent advances in the development of sigma receptor ligands as cytotoxic agents: A medicinal chemistry perspective. J. Med. Chem. 64, 7926–7962. doi: 10.1021/acs.jmedchem.0c02265,

PubMed Abstract | Crossref Full Text | Google Scholar

Feher, A., Juhasz, A., Laszlo, A., Kalman, J. Jr., Pakaski, M., Kalman, J., et al. (2012). Association between a variant of the sigma-1 receptor gene and Alzheimer's disease. Neurosci. Lett. 517, 136–139. doi: 10.1016/j.neulet.2012.04.046,

PubMed Abstract | Crossref Full Text | Google Scholar

Ferreira, S., Lourenco, M., Oliveira, M., and De Felice, F. (2015). Soluble amyloid-b oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front. Cell. Neurosci. 9:191. doi: 10.3389/fncel.2015.00191

Crossref Full Text | Google Scholar

Filadi, R., Greotti, E., Turacchio, G., Luini, A., Pozzan, T., and Pizzo, P. (2015). Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc. Natl. Acad. Sci. USA 112, E2174–E2181. doi: 10.1073/pnas.1504880112,

PubMed Abstract | Crossref Full Text | Google Scholar

Foretz, M., Guigas, B., Bertrand, L., Pollak, M., and Viollet, B. (2014). Metformin: from mechanisms of action to therapies. Cell Metab. 20, 953–966. doi: 10.1016/j.cmet.2014.09.018,

PubMed Abstract | Crossref Full Text | Google Scholar

Fujimoto, M., and Hayashi, T. (2011). New insights into the role of mitochondria-associated endoplasmic reticulum membrane. Int. Rev. Cell Mol. Biol. 292, 73–117. doi: 10.1016/B978-0-12-386033-0.00002-5,

PubMed Abstract | Crossref Full Text | Google Scholar

Giacomello, M., and Pellegrini, L. (2016). The coming of age of the mitochondria-ER contact: a matter of thickness. Cell Death Differ. 23, 1417–1427. doi: 10.1038/cdd.2016.52,

PubMed Abstract | Crossref Full Text | Google Scholar

Gómez-Suaga, P., Pérez-Nievas, B. G., Glennon, E. B., Lau, D. H. W., Paillusson, S., Mórotz, G. M., et al. (2019). The VAPB-PTPIP51 endoplasmic reticulum-mitochondria tethering proteins are present in neuronal synapses and regulate synaptic activity. Acta Neuropathol. Commun. 7:35. doi: 10.1186/s40478-019-0688-4,

PubMed Abstract | Crossref Full Text | Google Scholar

Granzotto, A., and Sensi, S. L. (2024). Once upon a time, the amyloid Cascade hypothesis. Ageing Res. Rev. 93:102161. doi: 10.1016/j.arr.2023.102161,

PubMed Abstract | Crossref Full Text | Google Scholar

Green, K. N. (2008). SERCA pump activity is physiologically regulated by presenilin and regulates amyloid beta production. J. Cell Biol. 181, 1107–1116. doi: 10.1083/jcb.200706171,

PubMed Abstract | Crossref Full Text | Google Scholar

Gregianin, E., Pallafacchina, G., Zanin, S., Crippa, V., Rusmini, P., Poletti, A., et al. (2016). Loss-of-function mutations in the SIGMAR1 gene cause distal hereditary motor neuropathy by impairing ER-mitochondria tethering and Ca2+ signalling. Hum. Mol. Genet. 25, 3741–3753. doi: 10.1093/hmg/ddw220,

PubMed Abstract | Crossref Full Text | Google Scholar

Grundman, M., Morgan, R., Lickliter, J. D., Schneider, L. S., DeKosky, S., Izzo, N. J., et al. (2019). A phase 1 clinical trial of the sigma-2 receptor complex allosteric antagonist CT1812, a novel therapeutic candidate for Alzheimer's disease. Alzheimer's & Dementia: Translational Res. Clin. Interventions 5, 20–26. doi: 10.1016/j.trci.2018.11.001,

PubMed Abstract | Crossref Full Text | Google Scholar

Guo, M., Liu, R., Zhang, F., Qu, J., Yang, Y., and Li, X. (2024). A new perspective on liver diseases: focusing on the mitochondria-associated endoplasmic reticulum membranes. Pharmacol. Res. 208:107409. doi: 10.1016/j.phrs.2024.107409,

PubMed Abstract | Crossref Full Text | Google Scholar

Ha, J., Choi, D. W., Kim, K. J., Cho, S. Y., Kim, H., Kim, K. Y., et al. (2021). Association of metformin use with Alzheimer's disease in patients with newly diagnosed type 2 diabetes: a population-based nested case-control study. Sci. Rep. 11:24069. doi: 10.1038/s41598-021-03406-5,

PubMed Abstract | Crossref Full Text | Google Scholar

Hampel, H., Hardy, J., Blennow, K., Chen, C., Perry, G., Kim, S. H., et al. (2021). The amyloid-beta pathway in Alzheimer's disease. Mol. Psychiatry 26, 5481–5503. doi: 10.1038/s41380-021-01249-0,

PubMed Abstract | Crossref Full Text | Google Scholar

Hampel, H., Williams, C., Etcheto, A., Goodsaid, F., Parmentier, F., Sallantin, J., et al. (2020). A precision medicine framework using artificial intelligence for the identification and confirmation of genomic biomarkers of response to an Alzheimer's disease therapy: analysis of the blarcamesine (ANAVEX2-73) phase 2a clinical study. Alzheimers Dement (NY) 6:e12013. doi: 10.1002/trc2.12013,

PubMed Abstract | Crossref Full Text | Google Scholar

Harper, A. R., Nayee, S., and Topol, E. J. (2015). Protective alleles and modifier variants in human health and disease. Nat. Rev. Genet. 16, 689–701. doi: 10.1038/nrg4017,

PubMed Abstract | Crossref Full Text | Google Scholar

Hayashi, T. (2019). The Sigma-1 receptor in cellular stress signaling. Front. Neurosci. 13:733. doi: 10.3389/fnins.2019.00733,

PubMed Abstract | Crossref Full Text | Google Scholar

Hayashi, T., and Su, T.-P. (2007). Sigma-1 receptor chaperones at the ER- mitochondrion Interface regulate Ca2+ signaling and cell survival. Cell 131, 596–610. doi: 10.1016/j.cell.2007.08.036,

PubMed Abstract | Crossref Full Text | Google Scholar

Hellewell, S. B., and Bowen, W. D. (1990). A sigma-like binding site in rat pheochromocytoma (PC12) cells: decreased affinity for (+)-benzomorphans and lower molecular weight suggest a different sigma receptor form from that of guinea pig brain. Brain Res. 527, 244–253.

Google Scholar

Hickman, L. B., Stern, J. M., Silverman, D. H. S., Salamon, N., and Vossel, K. (2023). Clinical, imaging, and biomarker evidence of amyloid- and tau-related neurodegeneration in late-onset epilepsy of unknown etiology. Front. Neurol. 14:1241638. doi: 10.3389/fneur.2023.1241638

Crossref Full Text | Google Scholar

Hirabayashi, Y., Kwon, S.-K., Paek, H., Pernice, W. M., Paul, M. A., Lee, J., et al. (2017). ER-mitochondria tethering by PDZD8 regulates Ca2+ dynamics in mammalian neurons. Science 358, 623–630. doi: 10.1126/science.aan6009,

PubMed Abstract | Crossref Full Text | Google Scholar

Huang, Y., Zheng, L., Halliday, G., Dobson-Stone, C., Wang, Y., Tang, H. D., et al. (2011). Genetic polymorphisms in sigma-1 receptor and apolipoprotein E interact to influence the severity of Alzheimer's disease. Curr. Alzheimer Res. 8, 765–770. doi: 10.2174/156720511797633232,

PubMed Abstract | Crossref Full Text | Google Scholar

Izzo, N. J., Xu, J., Zeng, C., Kirk, M. J., Mozzoni, K., Silky, C., et al. (2014). Alzheimer's therapeutics targeting amyloid beta 1–42 oligomers II: sigma-2/PGRMC1 receptors mediate Abeta 42 oligomer binding and synaptotoxicity. PLoS One 9:e111899. doi: 10.1371/journal.pone.0111899,

PubMed Abstract | Crossref Full Text | Google Scholar

Jansen, K. L., Faull, R. L., Storey, P., and Leslie, R. A. (1993). Loss of sigma binding sites in the CA1 area of the anterior hippocampus in Alzheimer's disease correlates with CA1 pyramidal cell loss. Brain Res. 623, 299–302. doi: 10.1016/0006-8993(93)91441-t,

PubMed Abstract | Crossref Full Text | Google Scholar

Ji, S., Zhao, X., Zhu, R., Dong, Y., Huang, L., and Zhang, T. (2022). Metformin and the risk of dementia based on an analysis of 396,332 participants. Ther Adv Chronic Dis 13:20406223221109454. doi: 10.1177/20406223221109454,

PubMed Abstract | Crossref Full Text | Google Scholar

Jin, J., Arbez, N., Sahn, J. J., Lu, Y., Linkens, K. T., Hodges, T. R., et al. (2022). Neuroprotective effects of σ(2)R/TMEM97 receptor modulators in the neuronal model of Huntington's disease. ACS Chem. Neurosci. 13, 2852–2862. doi: 10.1021/acschemneuro.2c00274,

PubMed Abstract | Crossref Full Text | Google Scholar

Jin, J. L., Fang, M., Zhao, Y. X., and Liu, X. Y. (2015). Roles of sigma-1 receptors in Alzheimer's disease. Int. J. Clin. Exp. Med. 8, 4808–4820.

Google Scholar

Katona, M., Bartok, A., Nichtova, Z., Csordas, G., Berezhnaya, E., Weaver, D., et al. (2022). Capture at the ER-mitochondrial contacts licenses IP(3) receptors to stimulate local ca(2+) transfer and oxidative metabolism. Nat. Commun. 13:6779. doi: 10.1038/s41467-022-34365-8,

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, M., and Bezprozvanny, I. (2023). Structure-based modeling of sigma 1 receptor interactions with ligands and cholesterol and implications for its biological function. Int. J. Mol. Sci. 24. doi: 10.3390/ijms241612980,

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, F. J., and Maher, C. M. (2017). “Sigma1 pharmacology in the context of Cancer” in Sigma proteins: Evolution of the concept of sigma receptors. eds. F. J. Kim and G. W. Pasternak (Cham: Springer International Publishing).

Google Scholar

Kowaltowski, A. J., Menezes-Filho, S. L., Assali, E. A., Gonçalves, I. G., Cabral-Costa, J. V., Abreu, P., et al. (2019). Mitochondrial morphology regulates organellar Ca2+ uptake and changes cellular Ca2+ homeostasis. FASEB J. 33, 13176–13188. doi: 10.1096/fj.201901136R,

PubMed Abstract | Crossref Full Text | Google Scholar

Leal, N. S., Dentoni, G., Schreiner, B., Naia, L., Piras, A., Graff, C., et al. (2020). Amyloid beta-peptide increases mitochondria-endoplasmic reticulum contact altering mitochondrial function and autophagosome formation in Alzheimer's disease-related models. Cells 9. doi: 10.3390/cells9122552

Crossref Full Text | Google Scholar

Leal, N. S., Schreiner, B., Pinho, C. M., Filadi, R., Wiehager, B., Karlstrom, H., et al. (2016). Mitofusin-2 knockdown increases ER-mitochondria contact and decreases amyloid beta-peptide production. J. Cell. Mol. Med. 20, 1686–1695. doi: 10.1111/jcmm.12863,

PubMed Abstract | Crossref Full Text | Google Scholar

Leuner, K., Muller, W. E., and Reichert, A. S. (2012). From mitochondrial dysfunction to amyloid beta formation: novel insights into the pathogenesis of Alzheimer's disease. Mol. Neurobiol. 46, 186–193. doi: 10.1007/s12035-012-8307-4,

PubMed Abstract | Crossref Full Text | Google Scholar

Leung, A., Ohadi, D., Pekkurnaz, G., and Rangamani, P. (2021). Systems modeling predicts that mitochondria ER contact sites regulate the postsynaptic energy landscape. NPJ Syst Biol Appl 7:26. doi: 10.1038/s41540-021-00185-7,

PubMed Abstract | Crossref Full Text | Google Scholar

Li, J., Félix-Soriano, E., Wright, K. R., Shen, H., Baer, L. A., Stanford, K. I., et al. (2022). Differential responses to Sigma-1 or Sigma-2 receptor ablation in adiposity, fat oxidation, and sexual dimorphism. Int. J. Mol. Sci. 23:10846. doi: 10.3390/ijms231810846,

PubMed Abstract | Crossref Full Text | Google Scholar

Luty, A. A., Kwok, J. B., Dobson-Stone, C., Loy, C. T., Coupland, K. G., Karlstrom, H., et al. (2010). Sigma nonopioid intracellular receptor 1 mutations cause frontotemporal lobar degeneration-motor neuron disease. Ann. Neurol. 68, 639–649. doi: 10.1002/ana.22274,

PubMed Abstract | Crossref Full Text | Google Scholar

Ma, T., Trinh, M. A., Wexler, A. J., Bourbon, C., Gatti, E., Pierre, P., et al. (2013). Suppression of eIF2α kinases alleviates Alzheimer's disease-related plasticity and memory deficits. Nat. Neurosci. 16, 1299–1305. doi: 10.1038/nn.3486,

PubMed Abstract | Crossref Full Text | Google Scholar

Macfarlane, S., Grimmer, T., Teo, K., O'Brien, T. J., Woodward, M., Grunfeld, J., et al. (2025). Blarcamesine for the treatment of early Alzheimer's disease: results from the ANAVEX2-73-AD-004 phase IIB/III trial. J. Prev Alzheimers Dis. 12:100016. doi: 10.1016/j.tjpad.2024.100016,

PubMed Abstract | Crossref Full Text | Google Scholar

Magalhaes Rebelo, A. P., Dal Bello, F., Knedlik, T., Kaar, N., Volpin, F., Shin, S. H., et al. (2020). Chemical modulation of mitochondria-endoplasmic reticulum contact sites. Cells 9, 1637–1655. doi: 10.3390/cells9071637,

PubMed Abstract | Crossref Full Text | Google Scholar

Malar, D. S., Thitilertdecha, P., Ruckvongacheep, K. S., Brimson, S., Tencomnao, T., and Brimson, J. M. (2023). Targeting sigma receptors for the treatment of neurodegenerative and neurodevelopmental disorders. CNS Drugs 37, 399–440. doi: 10.1007/s40263-023-01007-6,

PubMed Abstract | Crossref Full Text | Google Scholar

Martino Adami, P. V., Nichtova, Z., Weaver, D. B., Bartok, A., Wisniewski, T., Jones, D. R., et al. (2019). Perturbed mitochondria-ER contacts in live neurons that model the amyloid pathology of Alzheimer's disease. J. Cell Sci. 132. doi: 10.1242/jcs.229906,

PubMed Abstract | Crossref Full Text | Google Scholar

Maruszak, A., Safranow, K., Gacia, M., Gabryelewicz, T., Slowik, A., Styczynska, M., et al. (2007). Sigma receptor type 1 gene variation in a group of polish patients with Alzheimer's disease and mild cognitive impairment. Dement. Geriatr. Cogn. Disord. 23, 432–438. doi: 10.1159/000101990,

PubMed Abstract | Crossref Full Text | Google Scholar

Maurice, T., and Goguadze, N. (2017). Sigma-1 (sigma(1)) receptor in memory and neurodegenerative diseases. Handb. Exp. Pharmacol. 244, 81–108. doi: 10.1007/164_2017_15,

PubMed Abstract | Crossref Full Text | Google Scholar

Maurice, T., and Su, T. P. (2009). The pharmacology of sigma-1 receptors. Pharmacol. Ther. 124, 195–206. doi: 10.1016/j.pharmthera.2009.07.001,

PubMed Abstract | Crossref Full Text | Google Scholar

Megalizzi, V., Decaestecker, C., Debeir, O., Spiegl-Kreinecker, S., Berger, W., Lefranc, F., et al. (2009). Screening of anti-glioma effects induced by sigma-1 receptor ligands: potential new use for old anti-psychiatric medicines. Eur. J. Cancer 45, 2893–2905. doi: 10.1016/j.ejca.2009.07.011,

PubMed Abstract | Crossref Full Text | Google Scholar

Megalizzi, V., Mathieu, V., Mijatovic, T., Gailly, P., Debeir, O., De Neve, N., et al. (2007). 4-IBP, a sigma1 receptor agonist, decreases the migration of human cancer cells, including glioblastoma cells, in vitro and sensitizes them in vitro and in vivo to cytotoxic insults of proapoptotic and proautophagic drugs. Neoplasia 9, 358–369. doi: 10.1593/neo.07130,

PubMed Abstract | Crossref Full Text | Google Scholar

Merlos, M., Burgueno, J., Portillo-Salido, E., Plata-Salaman, C. R., and Vela, J. M. (2017). Pharmacological modulation of the sigma 1 receptor and the treatment of pain. Adv. Exp. Med. Biol. 964, 85–107. doi: 10.1007/978-3-319-50174-1_8,

PubMed Abstract | Crossref Full Text | Google Scholar

Misko, A. L., Sasaki, Y., Tuck, E., Milbrandt, J., and Baloh, R. H. (2012). Mitofusin2 mutations disrupt axonal mitochondrial positioning and promote axon degeneration. J. Neurosci. 32, 4145–4155. doi: 10.1523/JNEUROSCI.6338-11.2012,

PubMed Abstract | Crossref Full Text | Google Scholar

Morales-Lázaro, S. L., González-Ramírez, R., and Rosenbaum, T. (2019). Molecular interplay between the Sigma-1 receptor, steroids, and ion channels. Front. Pharmacol. 10:419. doi: 10.3389/fphar.2019.00419,

PubMed Abstract | Crossref Full Text | Google Scholar

Natsvlishvili, N., Goguadze, N., Zhuravliova, E., and Mikeladze, D. (2015). Sigma-1 receptor directly interacts with Rac1-GTPase in the brain mitochondria. BMC Biochem. 16:11. doi: 10.1186/s12858-015-0040-y,

PubMed Abstract | Crossref Full Text | Google Scholar

Nugent, A. A., Lin, K., van Lengerich, B., Lianoglou, S., Przybyla, L., Davis, S. S., et al. (2020). TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron 105, 837–854.e9. doi: 10.1016/j.neuron.2019.12.007,

PubMed Abstract | Crossref Full Text | Google Scholar

Oliveira, M. M., Lourenco, M. V., Longo, F., Kasica, N. P., Yang, W., Ureta, G., et al. (2021). Correction of eIF2-dependent defects in brain protein synthesis, synaptic plasticity, and memory in mouse models of Alzheimer's disease. Sci. Signal. 14. doi: 10.1126/scisignal.abc5429,

PubMed Abstract | Crossref Full Text | Google Scholar

Oyer, H. M., Steck, A. R., Longen, C. G., Venkat, S., Bayrak, K., Munger, E. B., et al. (2023). Sigma1 regulates lipid droplet-mediated redox homeostasis required for prostate Cancer proliferation. Cancer Res Commun 3, 2195–2210. doi: 10.1158/2767-9764.CRC-22-0371,

PubMed Abstract | Crossref Full Text | Google Scholar

Öztürk, Z., O’Kane, C. J., and Pérez-Moreno, J. J. (2020). Axonal endoplasmic reticulum dynamics and its roles in neurodegeneration. Front. Neurosci. 14. doi: 10.3389/fnins.2020.00048,

PubMed Abstract | Crossref Full Text | Google Scholar

Pao, P. C., Seo, J., Lee, A., Kritskiy, O., Patnaik, D., Penney, J., et al. (2023). A Cdk5-derived peptide inhibits Cdk5/p25 activity and improves neurodegenerative phenotypes. Proc. Natl. Acad. Sci. USA 120:e2217864120. doi: 10.1073/pnas.2217864120,

PubMed Abstract | Crossref Full Text | Google Scholar

Pera, M., Larrea, D., Guardia-Laguarta, C., Montesinos, J., Velasco, K. R., Agrawal, R. R., et al. (2017). Increased localization of APP-C99 in mitochondria-associated ER membranes causes mitochondrial dysfunction in Alzheimer disease. EMBO J. 36, 3356–3371. doi: 10.15252/embj.201796797,

PubMed Abstract | Crossref Full Text | Google Scholar

Piechal, A., Jakimiuk, A., and Mirowska-Guzel, D. (2021). Sigma receptors and neurological disorders. Pharmacol. Rep. 73, 1582–1594. doi: 10.1007/s43440-021-00310-7,

PubMed Abstract | Crossref Full Text | Google Scholar

Pilco-Ferreto, N., and Calaf, G. M. (2016). Influence of doxorubicin on apoptosis and oxidative stress in breast cancer cell lines. Int. J. Oncol. 49, 753–762. doi: 10.3892/ijo.2016.3558,

PubMed Abstract | Crossref Full Text | Google Scholar

Prinz, W. A., Toulmay, A., and Balla, T. (2020). The functional universe of membrane contact sites. Nat. Rev. Mol. Cell Biol. 21, 7–24. doi: 10.1038/s41580-019-0180-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Prudent, J., Zunino, R., Sugiura, A., Mattie, S., Shore, G. C., and McBride, H. M. (2015). MAPL SUMOylation of Drp1 stabilizes an ER/mitochondrial platform required for cell death. Mol. Cell 59, 941–955. doi: 10.1016/j.molcel.2015.08.001,

PubMed Abstract | Crossref Full Text | Google Scholar

Ray, B., Maloney, B., Sambamurti, K., Karnati, H. K., Nelson, P. T., Greig, N. H., et al. (2020). Rivastigmine modifies the α-secretase pathway and potentially early Alzheimer's disease. Transl. Psychiatry 10:47. doi: 10.1038/s41398-020-0709-x,

PubMed Abstract | Crossref Full Text | Google Scholar

Riad, A., Zeng, C., Weng, C.-C., Winters, H., Xu, K., Makvandi, M., et al. (2018). Sigma-2 receptor/TMEM97 and PGRMC-1 increase the rate of internalization of LDL by LDL receptor through the formation of a ternary complex. Sci. Rep. 8:16845. doi: 10.1038/s41598-018-35430-3,

PubMed Abstract | Crossref Full Text | Google Scholar

Ribeiro, F. C., Cozachenco, D., Heimfarth, L., Fortuna, J. T. S., de Freitas, G. B., de Sousa, J. M., et al. (2023). Synaptic proteasome is inhibited in Alzheimer's disease models and associates with memory impairment in mice. Commun Biol 6:1127. doi: 10.1038/s42003-023-05511-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Rishton, G. M., Look, G. C., Ni, Z.-J., Zhang, J., Wang, Y., Huang, Y., et al. (2021). Discovery of investigational drug CT1812, an antagonist of the Sigma-2 receptor complex for Alzheimer’s disease. ACS Med. Chem. Lett. 12, 1389–1395. doi: 10.1021/acsmedchemlett.1c00048,

PubMed Abstract | Crossref Full Text | Google Scholar

Rocha, A. G., Franco, A., Krezel, A. M., Rumsey, J. M., Alberti, J. M., Knight, W. C., et al. (2018). MFN2 agonists reverse mitochondrial defects in preclinical models of Charcot-Marie-tooth disease type 2A. Science 360, 336–341. doi: 10.1126/science.aao1785,

PubMed Abstract | Crossref Full Text | Google Scholar

Rountree, S. D., Atri, A., Lopez, O. L., and Doody, R. S. (2013). Effectiveness of antidementia drugs in delaying Alzheimer's disease progression. Alzheimers Dement. 9, 338–345. doi: 10.1016/j.jalz.2012.01.002,

PubMed Abstract | Crossref Full Text | Google Scholar

Ruiz-Cantero, M. C., Entrena, J. M., Artacho-Cordón, A., Huerta, M. Á., Portillo-Salido, E., Nieto, F. R., et al. (2024). Sigma-1 receptors control neuropathic pain and peripheral Neuroinflammation after nerve injury in female mice: A transcriptomic study. J. NeuroImmune Pharmacol. 19:46. doi: 10.1007/s11481-024-10144-8,

PubMed Abstract | Crossref Full Text | Google Scholar

Ruiz-Cantero, M. C., González-Cano, R., Tejada, M. Á., Santos-Caballero, M., Perazzoli, G., Nieto, F. R., et al. (2021). Sigma-1 receptor: A drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacol. Res. 163:105339. doi: 10.1016/j.phrs.2020.105339,

PubMed Abstract | Crossref Full Text | Google Scholar

Sałaciak, K., and Pytka, K. (2022). Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci. Biobehav. Rev. 132, 1114–1136. doi: 10.1016/j.neubiorev.2021.10.037,

PubMed Abstract | Crossref Full Text | Google Scholar

Sánchez-Blázquez, P., Cortés-Montero, E., Rodríguez-Muñoz, M., Merlos, M., and Garzón-Niño, J. (2020). The sigma 2 receptor promotes and the sigma 1 receptor inhibits mu-opioid receptor-mediated antinociception. Mol. Brain 13:150. doi: 10.1186/s13041-020-00676-4,

PubMed Abstract | Crossref Full Text | Google Scholar

Sanchez-Rangel, E., and Inzucchi, S. E. (2017). Metformin: clinical use in type 2 diabetes. Diabetologia 60, 1586–1593. doi: 10.1007/s00125-017-4336-x,

PubMed Abstract | Crossref Full Text | Google Scholar

Schmidt, H. R., and Kruse, A. C. (2019). The molecular function of σ receptors: past, present, and future. Trends Pharmacol. Sci. 40, 636–654. doi: 10.1016/j.tips.2019.07.006

Crossref Full Text | Google Scholar

Schmidt, H. R., Zheng, S., Gurpinar, E., Koehl, A., Manglik, A., and Kruse, A. C. (2016). Crystal structure of the human σ1 receptor. Nature 532, 527–530. doi: 10.1038/nature17391,

PubMed Abstract | Crossref Full Text | Google Scholar

Schon, E. A., and Area-Gomez, E. (2013). Mitochondria-associated ER membranes in Alzheimer disease. Mol. Cell. Neurosci. 55, 26–36. doi: 10.1016/j.mcn.2012.07.011,

PubMed Abstract | Crossref Full Text | Google Scholar

Shibuya, Y., Chang, C. C., Huang, L. H., Bryleva, E. Y., and Chang, T. Y. (2014). Inhibiting ACAT1/SOAT1 in microglia stimulates autophagy-mediated lysosomal proteolysis and increases Abeta1-42 clearance. J. Neurosci. 34, 14484–14501. doi: 10.1523/JNEUROSCI.2567-14.2014,

PubMed Abstract | Crossref Full Text | Google Scholar

Shibuya, Y., Niu, Z., Bryleva, E. Y., Harris, B. T., Murphy, S. R., Kheirollah, A., et al. (2015). Acyl-coenzyme A:cholesterol acyltransferase 1 blockage enhances autophagy in the neurons of triple transgenic Alzheimer's disease mouse and reduces human P301L-tau content at the presymptomatic stage. Neurobiol. Aging 36, 2248–2259. doi: 10.1016/j.neurobiolaging.2015.04.002,

PubMed Abstract | Crossref Full Text | Google Scholar

Silva, D. F., Selfridge, J. E., Lu, J., E, L., Cardoso, S. M., and Swerdlow, R. H. (2012). Mitochondrial abnormalities in Alzheimer's disease: possible targets for therapeutic intervention. Adv. Pharmacol. 64, 83–126. doi: 10.1016/B978-0-12-394816-8.00003-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Stacchiotti, A., Favero, G., Lavazza, A., Garcia-Gomez, R., Monsalve, M., and Rezzani, R. (2018). Perspective: mitochondria-ER contacts in metabolic cellular stress assessed by microscopy. Cells 8. doi: 10.3390/cells8010005,

PubMed Abstract | Crossref Full Text | Google Scholar

Su, T. P., Hayashi, T., Maurice, T., Buch, S., and Ruoho, A. E. (2010). The sigma-1 receptor chaperone as an inter-organelle signaling modulator. Trends Pharmacol. Sci. 31, 557–566. doi: 10.1016/j.tips.2010.08.007,

PubMed Abstract | Crossref Full Text | Google Scholar

Sugiura, A., Nagashima, S., Tokuyama, T., Amo, T., Matsuki, Y., Ishido, S., et al. (2013). MITOL regulates endoplasmic reticulum-mitochondria contacts via Mitofusin2. Mol. Cell 51, 20–34. doi: 10.1016/j.molcel.2013.04.023,

PubMed Abstract | Crossref Full Text | Google Scholar

Sukhorukov, V. S., Voronkova, A. S., Baranich, T. I., Gofman, A. A., Brydun, A. V., Knyazeva, L. A., et al. (2022). Molecular mechanisms of interactions between mitochondria and the endoplasmic reticulum: A new Look at how important cell functions are supported. Mol. Biol. 56, 59–71. doi: 10.1134/S0026893322010071

Crossref Full Text | Google Scholar

Tang, H., Tao, A., Song, J., Liu, Q., Wang, H., and Rui, T. (2017). Doxorubicin-induced cardiomyocyte apoptosis: role of mitofusin 2. Int. J. Biochem. Cell Biol. 88, 55–59. doi: 10.1016/j.biocel.2017.05.006,

PubMed Abstract | Crossref Full Text | Google Scholar

Terada, K., Migita, K., Matsushima, Y., Sugimoto, Y., Kamei, C., Matsumoto, T., et al. (2018). Cholinesterase inhibitor rivastigmine enhances nerve growth factor-induced neurite outgrowth in PC12 cells via sigma-1 and sigma-2 receptors. PLoS One 13:e0209250. doi: 10.1371/journal.pone.0209250,

PubMed Abstract | Crossref Full Text | Google Scholar

Trimmer, P. A., and Borland, M. K. (2005). Differentiated Alzheimer's disease transmitochondrial cybrid cell lines exhibit reduced organelle movement. Antioxid. Redox Signal. 7, 1101–1109. doi: 10.1089/ars.2005.7.1101,

PubMed Abstract | Crossref Full Text | Google Scholar

Tsunoda, T., Koga, H., Yokomizo, A., Tatsugami, K., Eto, M., Inokuchi, J., et al. (2005). Inositol 1,4,5-trisphosphate (IP3) receptor type1 (IP3R1) modulates the acquisition of cisplatin resistance in bladder cancer cell lines. Oncogene 24, 1396–1402. doi: 10.1038/sj.onc.1208313,

PubMed Abstract | Crossref Full Text | Google Scholar

Uchida, Y., Ohshima, T., Sasaki, Y., Suzuki, H., Yanai, S., Yamashita, N., et al. (2005). Semaphorin3A signalling is mediated via sequential Cdk5 and GSK3beta phosphorylation of CRMP2: implication of common phosphorylating mechanism underlying axon guidance and Alzheimer's disease. Genes Cells 10, 165–179. doi: 10.1111/j.1365-2443.2005.00827.x,

PubMed Abstract | Crossref Full Text | Google Scholar

van Dyck, C. H., Swanson, C. J., Aisen, P., Bateman, R. J., Chen, C., Gee, M., et al. (2023). Lecanemab in early Alzheimer's disease. N. Engl. J. Med. 388, 9–21. doi: 10.1056/NEJMoa2212948,

PubMed Abstract | Crossref Full Text | Google Scholar

Venkataraman, A. V., Mansur, A., Rizzo, G., Bishop, C., Lewis, Y., Kocagoncu, E., et al. (2022). Widespread cell stress and mitochondrial dysfunction occur in patients with early Alzheimer’s disease. Sci. Transl. Med. 14:eabk1051. doi: 10.1126/scitranslmed.abk1051,

PubMed Abstract | Crossref Full Text | Google Scholar

Voeltz, G. K., Sawyer, E. M., Hajnoczky, G., and Prinz, W. A. (2024). Making the connection: how membrane contact sites have changed our view of organelle biology. Cell 187, 257–270. doi: 10.1016/j.cell.2023.11.040,

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, T., and Jia, H. (2023). The sigma receptors in Alzheimer's disease: new potential targets for diagnosis and therapy. Int. J. Mol. Sci. 24. doi: 10.3390/ijms241512025,

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, M., Tian, T., Zhou, H., Jiang, S.-Y., Jiao, Y.-Y., Zhu, Z., et al. (2024). Metformin normalizes mitochondrial function to delay astrocyte senescence in a mouse model of Parkinson’s disease through Mfn2-cGAS signaling. J. Neuroinflammation 21:81. doi: 10.1186/s12974-024-03072-0,

PubMed Abstract | Crossref Full Text | Google Scholar

Watanabe, S., Ilieva, H., Tamada, H., Nomura, H., Komine, O., Endo, F., et al. (2016). Mitochondria-associated membrane collapse is a common pathomechanism in SIGMAR1- and SOD1-linked ALS. EMBO Mol. Med. 8, 1421–1437. doi: 10.15252/emmm.201606403,

PubMed Abstract | Crossref Full Text | Google Scholar

Weber, F., and Wünsch, B. (2017). Medicinal chemistry of σ(1) receptor ligands: pharmacophore models, synthesis, structure affinity relationships, and pharmacological applications. Handb. Exp. Pharmacol. 244, 51–79. doi: 10.1007/164_2017_33,

PubMed Abstract | Crossref Full Text | Google Scholar

Weng, T. Y., Tsai, S. A., and Su, T. P. (2017). Roles of sigma-1 receptors on mitochondrial functions relevant to neurodegenerative diseases. J. Biomed. Sci. 24:74. doi: 10.1186/s12929-017-0380-6,

PubMed Abstract | Crossref Full Text | Google Scholar

Wlodarczyk, J., Bhattacharyya, R., Dore, K., Ho, G. P. H., Martin, D. D. O., Mejias, R., et al. (2024). Altered protein Palmitoylation as disease mechanism in neurodegenerative disorders. J. Neurosci. 44:e1225242024. doi: 10.1523/JNEUROSCI.1225-24.2024,

PubMed Abstract | Crossref Full Text | Google Scholar

Xie, Q., Su, J., Jiao, B., Shen, L., Ma, L., Qu, X., et al. (2016). ABT737 reverses cisplatin resistance by regulating ER-mitochondria Ca2+ signal transduction in human ovarian cancer cells. Int. J. Oncol. 49, 2507–2519. doi: 10.3892/ijo.2016.3733,

PubMed Abstract | Crossref Full Text | Google Scholar

Yi, B., Sahn, J. J., Ardestani, P. M., Evans, A. K., Scott, L. L., Chan, J. Z., et al. (2017). Small molecule modulator of sigma 2 receptor is neuroprotective and reduces cognitive deficits and neuroinflammation in experimental models of Alzheimer's disease. J. Neurochem. 140, 561–575. doi: 10.1111/jnc.13917,

PubMed Abstract | Crossref Full Text | Google Scholar

Zellmer, J. C., Lomoio, S., Tanzi, R. E., and Bhattacharyya, R. (2025). Quantitative analysis of mitochondria-associated endoplasmic reticulum membrane (MAM) stabilization in a neural model of Alzheimer's disease (AD). JoVE :e66129. doi: 10.3791/66129,

PubMed Abstract | Crossref Full Text | Google Scholar

Zellmer, J. C., Tarantino, M. B., Kim, M., Lomoio, S., Maesako, M., Hajnóczky, G., et al. (2024). Stabilization of mitochondria-associated endoplasmic reticulum membranes regulates aβ generation in a three-dimensional neural model of Alzheimer's disease. Alzheimers Dement., 1–20. doi: 10.1002/alz.14417

Crossref Full Text | Google Scholar

Zhang, P., Konja, D., Zhang, Y., and Wang, Y. (2021). Communications between mitochondria and endoplasmic reticulum in the regulation of metabolic homeostasis. Cells 10. doi: 10.3390/cells10092195,

PubMed Abstract | Crossref Full Text | Google Scholar

Zhou, Y., Carmona, S., Muhammad, A., Bell, S., Landeros, J., Vazquez, M., et al. (2021). Restoring mitofusin balance prevents axonal degeneration in a Charcot-Marie-tooth type 2A model. J. Clin. Invest. 131. doi: 10.1172/JCI147307,

PubMed Abstract | Crossref Full Text | Google Scholar

Ziegler, D. V., Martin, N., and Bernard, D. (2021). Cellular senescence links mitochondria-ER contacts and aging. Commun Biol 4:1323. doi: 10.1038/s42003-021-02840-5,

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: sigma-1 and -2 receptors, σ1R/σ2R agonists and antagonists, amyotrophic lateral sclerosis, Alzheimer’s diseases, Huntington and Parkinson diseases, mitochondria-associated ER membrane

Citation: Lotlikar MS, Zellmer JC and Bhattacharyya R (2025) Sigma receptors and mitochondria-associated ER membranes are converging therapeutic targets for Alzheimer’s disease. Front. Neurosci. 19:1733659. doi: 10.3389/fnins.2025.1733659

Received: 27 October 2025; Revised: 22 November 2025; Accepted: 24 November 2025;
Published: 19 December 2025.

Edited by:

Zhi Dong Zhou, National Neuroscience Institute (NNI), Singapore

Reviewed by:

Tao Wang, North Sichuan Medical College, China
Kayalvizhi Rajendran, SASTRA University, India
Mustafa Shokr, Department of Pharmacology and Toxicology, Egypt

Copyright © 2025 Lotlikar, Zellmer and Bhattacharyya. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Raja Bhattacharyya, YmhhdHRhY2hhcnl5YS5yYWphQG1naC5oYXJ2YXJkLmVkdQ==

Present address: Madhura S. Lotlikar, Integrate Program of Neuroscience, McGill University, Québec, QC, Canada

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.