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

REVIEW article

Front. Neurosci., 15 December 2025

Sec. Neurodegeneration

Volume 19 - 2025 | https://doi.org/10.3389/fnins.2025.1665272

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

ER stress sensors at the ER-mitochondrial interface, controlling mitochondrial health in neurodegenerative diseases

Chaitanya Kunja&#x;Chaitanya KunjaVaishali Kumar&#x;Vaishali KumarPradeep KodamPradeep KodamChamundeshwari Devi GopuChamundeshwari Devi GopuShuvadeep Maity
Shuvadeep Maity*
  • Department of Biological Sciences, Birla Institute of Technology and Science (BITS)-Pilani (Hyderabad Campus), Hyderabad, Telangana, India

The endoplasmic reticulum (ER) and mitochondria are essential organelles that interact closely at specialized sites known as ER-mitochondria-associated membranes (MAMs). MAM is enriched with proteins from both the ER and mitochondria. ER stress sensors—inositol-requiring enzyme 1 (IRE1) and protein kinase RNA-like ER kinase (PERK) — are traditionally recognized for their roles in the unfolded protein response (UPR), which mitigates proteotoxic stress. However, recent studies reveal their non-canonical functions at MAMs, where they regulate calcium signaling, mitochondrial dynamics, and apoptosis through interactions with MAM-resident proteins. Disruption of these pathways is implicated in various diseases, particularly neurodegenerative disorders. This review highlights the emerging roles of IRE1 and PERK in preserving mitochondrial function and their relevance to neurodegeneration. It also examines pharmacological strategies targeting these proteins, which influence both UPR signaling and ER-mitochondrial communication, offering a comprehensive perspective on their roles in health and disease.

1 Introduction

In eukaryotic cells, the endoplasmic reticulum (ER) is the largest membrane-bound organelle, forming an extensive network of interconnected tubules and sheets. Based on the presence of membrane-bound ribosomes, the ER has been classified into two distinct types: rough ER and smooth ER. Together, these structures support a wide array of essential cellular functions, including protein and lipid synthesis, processing, and trafficking (Vance, 1990; Jacquemyn et al., 2017; Rahhali et al., 2023). Importantly, in coordination with mitochondria, the ER plays a pivotal role in calcium (Ca2+) storage and release, as well as in organelle biogenesis. This ER-mitochondrial communication is facilitated through a dynamic region termed the mitochondria-associated ER membrane (MAM). MAMs are enriched with transmembrane proteins that coordinate diverse signaling pathways (Janikiewicz et al., 2018; Missiroli et al., 2018). As a central hub for the synthesis of membrane and secretory proteins, the ER is particularly vulnerable to disruptions in protein folding, which may arise from environmental or pathological stressors. The accumulation of misfolded proteins triggers a condition known as ER stress, which activates a conserved signaling cascade termed the unfolded protein response of the ER (UPRER). ER transmembrane proteins, Inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like ER kinase (PERK), and Activating transcription factor 6 (ATF6) serve as primary sensors of ER stress and initiate the UPRER. Protein misfolding conditions can also activate mitochondrial UPR (UPRmt) primarily through activation of three transcription factors activating transcription factor 5 (ATF5), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP), which play crucial roles in maintaining mitochondrial health.

Interestingly, several studies have revealed that IRE1 and PERK are frequently localized at MAM, performing functions beyond their canonical roles in stress sensing (Verfaillie et al., 2012; Carreras-Sureda et al., 2019). In this review, we have discussed the non-canonical roles of IRE1 and PERK contributing towards different physiological processes other than UPRER. Further, we have described their implications in the pathogenesis of neurodegenerative diseases and reviewed therapeutic strategies targeting these proteins.

2 ER stress sensors at mitochondria-associated membranes (MAMs)

Mitochondria-associated membranes (MAMs) are specialized contact sites where the ER and mitochondria are closely tethered—within a distance of approximately 10–25 nm—by a set of bridging proteins, without complete membrane fusion. Technological advancements and accumulating evidence have revealed that MAMs function as intricate signaling hubs, composed of proteins derived from both the ER and the outer mitochondrial membrane (Csordás et al., 2006). During altered physiological conditions like ER and oxidative stress, the communication between the ER and mitochondria becomes vital for adapting to rapidly changing bioenergetic demands (Carreras-Sureda et al., 2019; Bassot et al., 2023). IRE1 and PERK regulate different signaling pathways in both canonical and non-canonical ways.

2.1 ER stress sensors activate their canonical signaling unfolded protein response (UPR) during ER stress

Typically, Inositol-requiring enzyme 1 (IRE1), encoded by the Ern1 gene (Endoplasmic Reticulum to Nucleus Signaling 1), is one of the most evolutionarily conserved transmembrane proteins of the ER and serves as a critical sensor of ER stress (Mori et al., 1993) (Figure 1). The canonical role of IRE1 was first identified in yeast through genetic screening, where it was recognized as a core component of the unfolded protein response (UPR) (Cox et al., 1993). In mammals, IRE1 exists in two isoforms: IRE1α and IRE1β. While budding yeast harbours only a single isoform, IRE1α is the predominant and ubiquitously expressed form in mammalian cells (Tirasophon et al., 1998). During its canonical response, IRE1α undergoes homodimerization after sensing misfolded protein accumulation inside the ER. Dimerization followed by autophosphorylation and subsequent conformational change activates its cytosolic RNase domain, which catalyses the unconventional splicing of HAC1 (in yeast) or XBP1 (in mammals) mRNA (Ron and Walter, 2007). The spliced form of XBP1 (or HAC1 in yeast) encodes a transcription factor that upregulates the transcription of UPR genes involved in protein folding, ER-associated degradation (ERAD), and lipid biosynthesis. In addition to these transcriptional responses, IRE1 also mediates regulated IRE1-dependent decay (RIDD) of select mRNAs, thereby reducing the protein folding burden on the ER and contributing to the restoration of proteostasis (Hollien and Weissman, 2006; Tam et al., 2014). Active IRE1α binds the adaptor protein TNFR-associated factor 2 (TRAF2), triggering the activation of the apoptosis signal-regulating kinase 1 (ASK1) and c-Jun/N-terminal kinase (JNK) pathway (Urano et al., 2000; Nishitoh et al., 2002).

Figure 1
Diagram showing the unfolded protein response (UPR) pathways. It details IRE1α dimerization/oligomerization leading to XBP1 splicing and RIDD, and PERK oligomerization causing eIF2α phosphorylation and selective translation of ATF4, inhibiting global translation. Outputs include UPR gene expression, ER chaperons, lipid synthesis, ERAD, and apoptosis via ASK and JNK.

Figure 1. Canonical pathway of ER stress activated by IRE1a and PERK in mammalian cells.: IRE1a undergoes dimerization oligomerization and autophosphorylation, leading to two main downstream events: (1) Unconventional splicing of XBP1u (unspliced) mRNA to produce spliced XBP1(XBP1s), which acts as an active transcription factor controlling UPR target gene expression [ER chaperons, Lipid biosynthesis, ER- associated degradation (ERAD)]. (2) interaction with TRAF2 to activate the JNK pathway mediated apoptosis via ASK1. PERK oligomerizes and auto phosphorylates, resulting in phosphorylation of elF2a, which inhibits global protein translation, allowing selective translation of ATF4. ATF4 functions as a transcription factor to induce genes involved in protein folding, and CHOP mediated apoptosis in case of prolonged stress.

PERK signaling is initiated by the phosphorylation at Ser51 of the α subunit of eukaryotic translation initiation factor 2 (eIF2α), resulting in a temporary cessation of global protein synthesis in response to ER stress or integrated stress response (ISR) (Figure 1). Interestingly, despite the overall inhibition of translation, a selective subset of translation remains active through stress-responsive transcription factors like ATF4, allowing the continued transcription of genes that regulate autophagy, ERAD, cell viability, and apoptosis (Almeida et al., 2022). ATF4 acts as a transcription factor of two key target genes, Growth Arrest and DNA damage 34 (GADD34) and C/EBP Homologous Protein (CHOP). GADD34, along with protein phosphatase 1 (PP1), dephosphorylates eukaryotic initiation factor 2α (eIF2α) to release the global translation arrest (Brush et al., 2003). On the other hand, CHOP can act as a rheostat that helps decide cellular fate (proliferation or apoptosis) during ER stress (Liu et al., 2024). CHOP can also control mitochondrial health as part of stress adaptation (Kaspar et al., 2021). The overlapping activation of PERK either via ER stress or ISR, can promote remodeling of mitochondria through both transcriptional as well as translational signaling (Almeida et al., 2022).

Beyond their canonical roles in detecting misfolded proteins and initiating the UPR, these proteins are thus increasingly recognized for their involvement in broader physiological functions mediated through ER-mitochondrial interactions (Figure 1). Here, we have focused on the ER-mitochondrial interactions governed by IRE1/PERK and other proteins to maintain cellular energy balance, redox status, and overall metabolic function.

2.2 IRE1—arm controlling ER-mitochondrial communication

In addition to its canonical function in the UPR, IRE1 plays a critical role in regulating different physiological processes, including ER-mitochondrial communication through its localization at MAM in various cellular contexts (Figure 2; Table 1).

Figure 2
Illustration of endoplasmic reticulum and mitochondrial interaction. Key components include IRE1α, Sig-1R, PERK, ERO1α, MFN1, and MFN2. Processes shown are calcium transfer, lipid transfer, and respiration. Non-canonical roles include cristae formation, dynamics maintenance, bioenergetic balance, and lipid transfer.

Figure 2. Non-canonical pathway activated by IRE1a and PERK: At MAM, IRE1a interacts with ion channel Sigma-1 receptor (Sig-1R) and inositol triphosphate receptor (IP3R), facilitating calcium transfer from the ER to mitochondria through VDAC-GRP75 complex, supporting mitochondrial respiration and ATP production. interaction of PERK with MFN1 and MFN2 contributes to ER-mitochondrial tethering and maintenance of morphology. The PERK-E-Syt1 complex involves in lipid transfer across membranes. Overall these interactions show non-canonical role of UPR sensors in the regulation of mitochondrial cristae formation, maintenance of dynamics, bioenergetic balance, and lipid homeostasis.

Table 1
www.frontiersin.org

Table 1. Non-canonical role of ER stress sensors IRE1 and PERK on organellar health.

Mori et al., 2013 reported that IRE1α is highly enriched at MAM in Chinese hamster ovary (CHO) cells. In this study, the authors identified the Sigma-1 receptor (Sig-1R), a calcium-sensitive and ligand-regulated chaperone, as a crucial component of this interface. Under normal physiological conditions, Sig-1R forms a complex with the ER chaperone BiP. However, Sig-1R dissociates from BiP upon ER calcium depletion and stabilizes inositol 1,4,5-trisphosphate receptors (IP3Rs), facilitating sustained calcium signaling from the ER to mitochondria. This was the first demonstration of an ER-resident chaperone directly modulating mitochondrial calcium signaling. Importantly, another study found Sig-1R also interacts physically with IRE1α at MAMs under ER stress conditions. This interaction is reactive oxygen species (ROS)-dependent and contributes to the stabilization of IRE1α at the MAM, thereby prolonging the IRE1-XBP1 signaling axis and promoting cell survival during stress (Mori et al., 2013).

Further insights into the regulatory role of IRE1α were provided by Takeda et al. (2019), who demonstrated that mitochondrial ubiquitin ligase MITOL (also known as MARCH 5) prevents ER stress-induced apoptosis by ubiquitylating IRE1α at MAM. MITOL-mediated K63-linked chain ubiquitination of IRE1α suppresses IRE1α hyper-oligomerization, thereby limiting its pro-apoptotic signaling and reducing regulated IRE1-dependent decay (RIDD) during ER stress (Takeda et al., 2019). Here, MAM serves as a signaling site for this protective adaptation, helping fine-tune IRE1α’s activity under stress conditions.

Expanding on the non-canonical functions of IRE1α, Carreras-Sureda et al. reported that conditional knockout of IRE1α in mouse liver resulted in significant alteration of mitochondrial metabolism. Interestingly, the study pointed out a reduction of MAM proteins but not mitochondrial proteins. In vitro studies with combinations of different mutants of IRE1α further revealed that a direct interaction between IRE1α and IP3R1 is essential for mitochondrial calcium uptake (Carreras-Sureda et al., 2019). These findings highlight the physiological importance of IRE1α in maintaining ER-mitochondrial homeostasis beyond its classical role in the UPR.

The role of IRE1α in maintaining mitochondrial integrity has also been demonstrated in podocytes or Glomerular epithelial cells (GEC). Navarro-Betancourt et al. showed that podocyte-specific knockout of IRE1α in mice led to significant mitochondrial dysfunction. These cells exhibited reduced respiratory capacity, diminished mitochondrial membrane potential, and structural abnormalities. Electron microscopy revealed loss of cristae, increased mitochondrial roundness, and decreased matrix density—hallmarks of impaired mitochondrial health. These findings underscore the essential role of IRE1α in preserving mitochondrial structure and function, particularly in metabolically active cells (Navarro-Betancourt et al., 2020).

The IRE1α–XBP1 axis has also been shown to regulate metabolic flexibility in immune cells. In macrophages, activation of this pathway is essential for the metabolic shift toward glycolysis following lipopolysaccharide (LPS) infection. This glycolytic switch enables rapid ATP production, which is critical for sustaining the pro-inflammatory response. Conditional knockout of IRE1α or XBP1 in macrophages impaired the expression of glycolytic genes, disrupted glycolytic flux, and compromised mitochondrial metabolism. As a result, energy production was insufficient to maintain immune activation, highlighting the axis’s role in coordinating energy metabolism during immune stress (English et al., 2023a).

In the neuronal system, the IRE1α-XBP1 signaling axis plays a pivotal role at MAMs, where it helps maintain ER-mitochondrial tethering and supports mitochondrial function. A recent study demonstrated that treatment of SH-SY5Y neuroblastoma cells with Aβ₍₂₅₃₅₎ activates the IRE1–XBP1 pathway, enhancing interactions with key MAM proteins such as IP3R, GRP75, and VDAC1. This leads to increased calcium transfer from the ER to mitochondria. However, excessive calcium influx can result in mitochondrial dysfunction. Notably, inhibition of the IRE1–XBP1 axis restored mitochondrial function and reduced MAM-associated interactions, suggesting that this pathway fine-tunes ER-mitochondrial communication to preserve mitochondrial health under stress conditions (Chu et al., 2021).

IRE1 which typically activates pro-survival signaling pathways can induce apoptosis for prolonged ER stress. Due to its ability to be involved in two opposing functions, the activation of inhibition of this pathway leads to different effects based on the physiological condition of the cells. For example, chronic ER stress has been associated with retinal degeneration and diabetes. Thus, small molecule’s targeted inhibition of IRE1α was protective against retinal degeneration in the rat model and against pancreatic β-cells in the Akita diabetic mouse model (Ghosh et al., 2014). Interestingly, not the ER stress pathways but IRE1α knockdown has been shown to induce cell death in SH-SY5Y by disrupting intracellular Ca2 + homeostasis (Son et al., 2014). The RIDD via UPR sensor IRE1α transiently catalyzed DR5 mRNA decay. This allows time for adaptation during chronic ER stress, promoting survival by inhibiting pro-apoptotic proteins like DR5 (Lu et al., 2014). Using Drosophila model of PD Yan et al. found that IRE1 promotes neuronal death of dopaminergic neurons and locomotor impairment through activating autophagic pathways. Thus, inhibiting IRE1 or its downstream autophagic proteins (ATG7) ameliorated the progression of α-synuclein-caused Parkinson’s disease (Yan et al., 2019; Hetz et al., 2020; Kim et al., 2022). On the contrary, genetic disruption of the ER stress sensor IRE1 accelerated age-related cognitive decline. In the context of brain aging, a mouse brain study demonstrated that impairment of UPR signaling begins with age. Over-expression XBP1s or XBP1s gene delivery to aged mice not only improved age associated phenotypes but also reverted brain dysfunction, indicating sustained expression of IRE1α-XBP1 pathway is essential to protect against age and age related disorders (Cabral-Miranda et al., 2022a). Further, the involvement of the IRE1-PERK arm as a part of MAM in controlling neurological disorders is discussed in a separate section. Collectively, these findings establish the IRE1–XBP1 axis as a key regulator of cellular survival, which also coordinates with other pathways, including mitochondrial function, depending on various cellular contexts.

2.3 PERK—arm controlling ER-mitochondrial communication

PERK maintains mitochondrial homeostasis, mitochondrial cristae junctions, and bioenergetics depending on the needs of cells. During ER stress, a discordant behavior of transcription and translation was observed by Rendleman et al. (2018). An in-depth integrated multi-omics study pointed out that even during the halt of the global translation, a subset of genes supports the rerouting of energy production via upregulated translation, but not transcription, of Complexes I-IV from the oxidative phosphorylation pathway. At the same time, Lebeau et al. (2018) pointed out stress-induced hyperfusion of mitochondria through PERK-mediated signaling in coordination with the inner membrane protease YME1L. The use of IRE1, PKA, HDAC, and PERK inhibitors revealed that YME1L supports mitochondrial elongation via PERK signaling during ER stress. A follow-up study showed that PERK regulates phosphatidic acid (PA) levels at the outer mitochondrial membrane by promoting YME1L-dependent degradation of the PA transporter PRELID. This mechanism facilitates mitochondrial fusion, highlighting PERK’s critical role in mitochondrial dynamics under stress (Perea et al., 2023). Contemporary research has shed light on the metabolic adaptation of mitochondria during early ER stress via ERO1α. At the early phase of ER stress, PERK interacts with oxidoreductase ERO1α, followed by the formation of a heterocomplex (PERK-ERO1⍺) through the C-terminal active site of ERO1⍺ and cysteine 216 of PERK. This redox-dependent complex with ERO1α promotes MAM protein oxidation and enhances ER-mitochondrial Ca2+ flux. An earlier interactome analysis revealed that PERK aids Ca2+ signaling by interacting with the SERCA pump (Sassano et al., 2021). These interactions help to maintain mitochondrial dynamics and bioenergetic balance while limiting oxidative stress (Bassot et al., 2023). PERK deficient cells showed altered vulnerability during ROS-mediated ER stress following ROS signaling between the ER and mitochondria (Verfaillie et al., 2012).

PERK interacts with the actin-binding protein Filamin A (FLNA), which plays a key role in F-actin remodelling. This PERK–FLNA axis is essential for expanding ER–plasma membrane (PM) contacts following ER Ca2+ depletion. Loss of PERK disrupts F-actin organization, alters the F/G actin ratio, and reduces cell motility. Notably, this activity is driven by Ca2+ signaling and occurs independently of classical UPR pathways. These findings further underscore PERK’s critical role in regulating mitochondrial dynamics and cytoskeletal integrity (Muñoz et al., 2013). PERK also plays a key role as a regulator of lipid trafficking at ER-mitochondria contact sites through its interaction with the lipid transfer protein E-Syt1. Disruption of this PERK–E-Syt1 complex impairs lipid transfer and compromises mitochondrial respiratory capacity (Sassano et al., 2023). During cold stress or high energy demand, PERK facilitates mitochondrial cristae formation and respiration by activating O-linked N-acetylglucosamine transferase (OGT), which glycosylates TOM70 at Ser94 and enhances MIC19 import in mitochondria (Latorre-Muro et al., 2021).

Fusion proteins, Mitofusin 1 (MFN1) and Mitofusin 2 (MFN2), are found to interact with PERK. Mfn2 can act as an upstream modulator of PERK. Ablation of Mfn2 causes sustained kinase activation of PERK. In a healthy condition, Mfn2 keeps PERK in an inactive state. An altered environment, such as a high glucose level, causes activation of PERK after a reduction in the Mfn2 level and a disturbance of MAM signaling with subsequent mitochondrial dysfunction (Muñoz et al., 2013).

Mitochondrial thermogenesis in brown adipose tissue is also regulated by the ER-resident UPR sensor PERK. During brown adipocyte differentiation, PERK becomes phosphorylated independently of ER stress. This activation enhances the transcriptional activity of GABPα, which is essential for mitochondrial inner membrane protein biogenesis. Although GABPα protein expression remains unchanged in PERK-deficient cells, its transcriptional activity is significantly reduced—an effect not observed in IRE1α- or ATF6-deficient cells—highlighting a unique PERK–GABPα signaling axis that may be targeted to boost energy metabolism without disrupting ER homeostasis (Kato et al., 2020).

Beyond controlling mitochondria, PERK plays a critical role in inflammation. The nucleic acid sensor cGAS detects cytosolic DNA from invading pathogens and synthesizes the secondary messenger 2′3′-cGAMP. This molecule binds to the ER-resident adaptor protein STING, a key player in innate immunity. STING engages PERK via its intracellular domain upon activation, triggering non-canonical activation of PERK. Activated PERK then phosphorylates eIF2α, initiating translational programs that support inflammatory and survival responses (Zhang et al., 2024).

3 IRE1/PERK at the interface of neurodegenerative disease

Neurodegenerative diseases, characterized by the progressive dysfunction and loss of neurons, are increasingly linked to disruptions of cellular proteostasis, particularly in the ER (Tam et al., 2014). As mentioned earlier, PERK, plays a pivotal role in maintaining cellular homeostasis under ER stress conditions (Shacham et al., 2021). However, under chronic ER stress, sustained PERK activation can shift from protective to deleterious, contributing to neuronal dysfunction and death (Xu et al., 2017). Increased levels of phosphorylated PERK (p-PERK) and phosphorylated eIF2α (p-eIF2α) were found in the brains of patients with neurodegenerative diseases, suggesting that ER stress and PERK activation may play a role in disease pathogenesis (Bell et al., 2016). Indeed, PERK activation has been directly linked to synaptic dysfunction, impaired long-term potentiation, and cognitive deficits. A significant example of PERK’s role in neurodegeneration comes from a study on tauopathies — a class of neurodegenerative diseases marked by tau aggregation. Park et al. elucidated that PERK (EIF2AK3) helps in preventing tau aggregation. Using HEK293 cells expressing TauRD (P301S)-YFP and PERK+/+ and PERK−/− mouse embryonic fibroblasts, authors demonstrated that PERK activation mitigates tau aggregation, while its inhibition exacerbates pathology. Studies on tauopathy patient brain tissue exhibited a correlation between diminished PERK signaling and increased tau pathology, underscoring the protective role of PERK against tau aggregation and suggesting a potential therapeutic target, particularly in individuals with tauopathy-associated PERK hypomorph alleles (Park et al., 2023). Conversely, downregulation of PERK was found to be beneficial for neurodegenerative conditions. Wang et al. demonstrated that PERK downregulation protects rat Müller glial cells under oxygen–glucose deprivation (OGD) (Wang et al., 2023). Furthermore, OGD-induced phosphorylated tau (p-Tau) expression in rMC-1 cells was diminished by PERK knockout, implicating PERK’s role in tau-related pathology under stress. PERK knockout reduced the expression of autophagy-related proteins (LC3 and Beclin1) and apoptotic markers, improving cellular viability. PERK can reduce tau phosphorylation and autophagic stress, offering protective benefits to retinal neurons and glia (Chiu et al., 2023). Interestingly, a recent study found that the selective removal of astrocytic PERK decreases toxic aggregation of β-amyloid and tau proteins with enhanced glymphatic clearance and improved cognitive performance in mice (Chen et al., 2025). In Alzheimer’s disease (AD), IRE1 activation has been correlated with increased amyloid-beta (Aβ) production and tau phosphorylation, thereby exacerbating neurodegenerative processes. Notably, activation of the IRE1α/TRAF2/JNK signaling pathway has been implicated in neuronal apoptosis across several neurodegenerative disorders (Wan et al., 2020). Targeting PERK and IRE1 presents a promising therapeutic strategy given their wide-reaching impact. Genetic or pharmacological modulation of IRE1 can significantly affect cell viability (Siwecka et al., 2021). Ablation of IRE1’s RNase domain reduces amyloid deposits, Aβ oligomer levels, and astrocyte activation in the brain of a mouse model (Duran-Aniotz et al., 2017). A very recent study identified a novel non-canonical function of the IRE1α–Xbp1 branch in regulating the proteostasis of poly (GR) via preventing its accumulation in Amyotrophic Lateral Sclerosis (ALS). Using different cellular and in vivo models authors demonstrated that ectopic expression of IRE1 or its downstream effector XBP1s or pharmacological activation of IRE1 significantly reduced poly (GR) protein levels and ameliorated associated proteotoxic phenotype (Li et al., 2025a).

Huntington’s disease (HD) is another condition where sustained UPR activation contributes to disease pathology. Mutant huntingtin protein (mHTT) leads to chronic ER stress in HD models, with upregulation of markers such as GRP78 and CHOP observed in the hippocampus even at early disease stages. In R6/1 HD mice, ER stress appears to be mediated by PERK hyperactivation, which impairs cognition through prolonged suppression of essential protein synthesis for plasticity and memory. Notably, genetic reduction of GRP78 expression mitigates spatial and recognition memory deficits, suggesting targeted therapeutic intervention can be achieved by modulating these pathways (Shacham et al., 2021; Espina et al., 2023).

Further evidence of the protective role of IRE1 comes from studies involving parkin overexpression. Both in vitro and in vivo studies demonstrated that increased XBP1s levels through overexpression of parkin potentially confer resistance to PD-related toxins in dopaminergic neurons. In PD, cell survival has been shown to be linked with nitric oxide (NO) exposure. Studies using parkin and its pathogenic mutants in SH-SY5Y neuroblastoma cells identified the protective role of the IRE1α/XBP1 signaling axis against NO-induced apoptosis. Parkin also enhances the activity of the RNase domain of IRE1α following treatment with nitric oxide (NO) donor, S-nitroso-N-acetyl penicillamine (SNAP). The protective activity of parkin was found to be compromised after pharmacological inhibition or genetic knockdown of the RNase activity. Pathogenic parkin mutants showed a lower ability to activate the IRE1α/XBP1 signaling (Chiu et al., 2023). In Marinesco-Sjögren syndrome (MSS), PERK inhibition delays neurodegeneration and improves motor function (Grande et al., 2018).

Beyond neurodegenerative diseases, multiple studies have consistently highlighted the connection between ER stress. ER stress markers are also dysregulated in neuropsychiatric disorders. For instance, Kim et al. and Xue et al., demonstrated altered expression of BiP, PERK, and XBP1 in the prefrontal cortex, along with elevated serum levels of ATF6 and XBP1 in individuals with schizophrenia. These molecular signatures suggest that ATF6 and XBP1 may serve as biomarkers and functional contributors to schizophrenia. Similarly, individuals with bipolar disorder (BD) show an impaired UPR under ER stress. A specific SNP (–116C → G) in the XBP1 promoter has been associated with increased BD risk, while lymphoblasts from BD patients exhibit reduced expression of UPR genes such as XBP1 and CHOP under ER stress conditions (Kim et al., 2021; Xue et al., 2023). In the pathophysiology of Major Depressive Disorder (MDD), elevated levels of ER stress markers such as GRP78, CHOP, and XBP1 have been found in both animal models and human subjects with depression. Notably, activation of the PERK-eIF2α pathway in the hippocampus is associated with decreased brain-derived neurotrophic factor (BDNF) levels and depression-like behavior. Interestingly, inhibiting PERK improves memory performance, emphasizing its role in cognition (Sharma et al., 2018). Elevated expression of ER stress-related genes has also been reported in learned helplessness models and post-mortem brain tissue from MDD patients, especially those who died by suicide. Furthermore, peripheral blood cells of MDD patients exhibit heightened ER stress markers, further affirming the role of ER stress in MDD pathophysiology (Nevell et al., 2014; Behnke et al., 2016).

The role of UPR extends beyond pathology to developmental processes. During early brain development, UPR activation, particularly via IRE1 and PERK pathways, significantly influences neurogenesis. Mouse embryonic stem cell models have shown that UPR induction promotes neuronal over glial differentiation, underscoring its impact on lineage specification. The PERK/ATF4 signaling axis is critical for neurogenesis and proper neuron positioning in the developing cortex (Laguesse et al., 2015). However, excessive ER stress during cortical development, such as that induced by reduced codon translation rates, may skew neurogenesis toward direct pathways, potentially leading to microcephaly.

UPR elements also regulate neurogenesis and synaptic connectivity. Specifically, BDNF-induced UPR signaling in neurons is essential for its outgrowth. XBP1 deficiency impairs neurite growth indicating the activation of IRE1α-xbp1 is essential in developing central nervous system (CNS) of mammals (Hayashi and Su, 2007; Edvardson et al., 2019). This intricate regulation of synaptic function by the UPR is evident in various neurodegenerative disorders. Phosphorylation of eIF2α plays a central role in the synaptic dysfunction and memory impairments observed in Alzheimer’s, tauopathies, and prion diseases. Reducing p-eIF2α levels or modulating its upstream kinases (e.g., PERK, GCN2, PKR) has improved synaptic plasticity and memory in animal models (Smith and Mallucci, 2016). Moreover, XBP1s enhance cognitive function by regulating BDNF and dendritic spine-associated proteins, indicating the broader implications of UPR components in neuropsychiatric and neurodegenerative disorders (Gerakis and Hetz, 2018). A study on mouse brain aging demonstrated that impaired UPR signaling begins by middle age and ablation of IRE1 expression accelerates age-associated cognitive decline (Cabral-Miranda et al., 2022).

MAM further complicates the ER stress signaling in pathological conditions. The close juxtaposition of the ER and mitochondria at MAMs enables efficient calcium ion transfer from ER to mitochondria, which is essential for mitochondrial ATP production, buffering of cytosolic calcium levels, and modulation of the intrinsic apoptotic pathway (Hetz et al., 2020; Kim et al., 2022). Dysfunction of MAM components and the resulting alterations in calcium signaling have been implicated in several neurodegenerative conditions, highlighting the critical interplay between PERK, IRE1, and MAM-resident proteins in maintaining neuronal integrity (Corazzari et al., 2017). The therapeutic intervention also includes the modulation of MAM proteins, sigma-1 receptor (Sig-1R), and IRE1α. Tan et al. (2024) demonstrated that activating the sigma-1 receptor enhanced motor function recovery, reduced apoptosis, and ameliorated neuronal ferroptosis via IRE1α.

4 Pharmacological intervention targeting IRE1 and PERK

Till now, we have described the involvement of IRE1/PERK in shaping inter-organellar communication and its participation in the neurodegenerative diseases. Due to their involvement in various physiological and pathophysiological processes, a careful pharmacological intervention could be one of the best strategies to combat diseases. A recent study elucidated the crosstalk between the IRE1 and PERK branches of UPR during prolonged ER stress through pharmacological inhibition. The authors demonstrated that IRE1-XBP1s signaling is crucial in sustaining PERK expression under chronic stress conditions. Treatment with various IRE1 inhibitors, including MKC8866, reduced PERK signaling during extended ER stress, suggesting that these inhibitors may also suppress PERK expression and activity. Furthermore, the study shows that IRE1’s RNase activity enhances PERK transcription through the IRE1-XBP1s axis. These findings highlight that the IRE1, PERK, and ATF6 pathways function as an interconnected signaling network, rather than as isolated, parallel branches of the UPR (Ong et al., 2024). IRE1 inhibitors can work by inhibiting both the kinase and the RNase domain or blocking either the kinase or the RNase domain individually. Thus, these inhibitors block IRE1 phosphorylation-driven signaling cues or IRE1 signaling activated by the RNase domain (e.g., RIDD and XBP1 splicing). This specific activity provides a useful strategy to target the required signaling depending on the disease.

Inhibitors like 4μ8c and STF-083010 have been found effective against cancer cell proliferation, demonstrating the potential of inhibiting pro-survival UPR functionality by blocking RIDD (Papandreou et al., 2011; Cross et al., 2012). Kinase domain-specific inhibitor of IRE1α, kinase-inhibiting RNase attenuators 6 (KIRA6), an ATP-competitive inhibitor, disrupts IRE1α oligomerization, reducing RNase activity and promoting cell survival under stress. Kinase domain-specific inhibitor of IRE1α shows therapeutic potential in conditions like retinal degeneration and hyperglycemia by protecting photoreceptors and pancreatic β cells (Cross et al., 2012; Guirao-Abad et al., 2020). With respect to multiple myeloma (MM) treatment, bortezomib (BTZ), a proteasome inhibitor, is often found to be effective for very short duration. The drug resistance tends to develop rapidly against BTZ. To overcome the limitation and in search of new therapeutics, Ri et al. used toyocamycin, an XBP1 inhibitor derived from an Actinomycete strain. Toyocamycin selectively inhibits the IRE1α-XBP1 pathway by preventing XBP1 mRNA cleavage without activating ATF6 or PERK signaling. It blocks both constitutive and ER stress-induced XBP1 mRNA splicing and induces apoptosis even in bortezomib-resistant MM cells. Additionally, toyocamycin with BTZ showed a synergistic effect over MM (Ri et al., 2012).

Dual inhibitors, such as MKC-3946 and GSK2850163, are also effective in cancer treatment by modulating kinase-driven XBP1 splicing and RNase-driven mRNA decay, offering therapeutic potential for chronic ER stress-related diseases. MKC-3946 enhances CHOP-mediated apoptosis and blocks XBP1 splicing induced by chemotherapeutic agents, making it a promising candidate for improving cancer treatment outcomes, particularly in MM. The above-described inhibitors are mainly effective against different cancers where ER stress sensors play a significant role in cell survival. Interestingly, while inhibiting IRE1 function, with these inhibitors also alter mitochondrial activity, indicating disruption of ER-mitochondrial crosstalk.

STF-083010, an inhibitor of IRE1α endonuclease activity, significantly impacts mitochondrial functions, including decreased maximal mitochondrial respiration and spare respiratory capacity. It has also been observed that it can alter mitochondrial membrane potential and increase mitochondrial sensitivity to uncouplers. Importantly, these effects are independent of inhibiting IRE1α endonuclease activity, possibly due to STF-083010’s hydrophobic nature, which allows it to accumulate in cell membranes and directly act on mitochondrial dynamics (Hatokova et al., 2023).

Another IRE1α inhibitor, 4μ8c, plays an important role in modulating ER-mitochondria crosstalk. Aβ treatment has been shown to increase the expression of MAM proteins such as IP3R, Grp75, and VDAC1, leading to increased ER-mitochondria connection and then damage of MAMs and mitochondrial dysfunction. Pre-treatment with 4μ8c rescued the length of ER-mitochondria contact sites and also restored calcium homeostasis. Similarly, APY29 significantly affects ER-mitochondria crosstalk by disrupting cellular homeostasis and inducing apoptosis in ovarian granulosa cells (SVOG). APY29 has shown to intensify reactive oxygen species (ROS) and also decrease mitochondria membrane potential significantly; both are critical indicators of mitochondrial health and function (Li et al., 2025b).

PERK inhibitors essentially block ER stress as well as ISR pathways. The ISR involves four key kinases, with PERK playing a central role. Upon ER stress (e.g., thapsigargin exposure), PERK phosphorylates eIF2α, suppressing global protein synthesis while promoting translation of adaptive genes like ATF4. This enhances mitochondrial proteostasis, increases phosphatidic acid on the outer mitochondrial membrane, inhibits the pro-fission protein DRP1, and promotes mitochondrial elongation—strengthening ER-mitochondria contact. ISRIB, an ISR inhibitor, counteracts eIF2α phosphorylation to test whether these mitochondrial changes are PERK-dependent (Baron et al., 2025).

GSK2606414 is a selective PERK inhibitor against high glucose-induced neurotoxicity in mouse neuroblastoma cells. It attenuates the UPR by downregulating ER stress markers such as GRP78, p-PERK, p-eIF2α, ATF4, and CHOP. This inhibition also reduces pro-apoptotic proteins Bax and caspase-3 while increasing anti-apoptotic Bcl-2 levels, highlighting its potential in mitigating ER stress-related neuronal damage (Gundu et al., 2022). GSK2606414 also enhances mitochondrial function by restoring levels of TFAM, NRF1, Complex I, Complex II, and ATP synthase subunit C. It also reverses the downregulation of MFN2, a key ER-mitochondria tethering protein, and reduces mitochondrial superoxide accumulation (Gundu et al., 2022).

In prion-infected mice, GSK2606414 demonstrates strong neuroprotective effects by inhibiting PERK phosphorylation, reducing p-eIF2α, ATF4, and CHOP levels, restoring global protein synthesis, and preserving synaptic proteins like SNAP25 and PSD95 (Moreno et al., 2013). However, at micromolar concentrations, GSK2606414, along with GSK2656157 and AMG44, can off-target activate the ISR via GCN2 due to increased ATP affinity at its kinase domain. This dose-dependent activation occurs without ER stress, underscoring the importance of precise dosing when using PERK inhibitors to study ISR-driven mitochondrial remodelling (Szaruga et al., 2023).

GSK2656157, another PERK inhibitor, exhibits neuroprotective effects in a subarachnoid haemorrhage (SAH) rat model by modulating ER stress and suppressing TXNIP signaling. Following SAH, TXNIP expression increases in neurons, microglia, and astrocytes, contributing to early brain injury via pro-inflammatory and pro-apoptotic pathways. Intra-cerebroventricular administration of GSK2656157 inhibits PERK autophosphorylation and downregulates ER stress markers (GRP78, p-PERK, p-eIF2α, ATF4, CHOP), reducing inflammation and apoptosis. It also restores mitochondrial function by increasing the expression of TFAM, NRF1, Complex I, Complex II, and ATP synthase subunit C (Yan et al., 2017). Like inhibitors, PERK activators are also effective therapeutics in certain neurodegenerative conditions. Studies of cellular and mouse models of tauopathy demonstrated that treatment of CCT020312 improved memory and motor function via reduction of tau phosphorylation in P301S tau mice (Bruch et al., 2017). Similarly, another PERK activator, MK28, reduced ER stress-induced apoptosis in a striatal cell line (STHdhQ111/111) while in vivo administration improved motor function and life span of the R6/2 HD mice (Ganz et al., 2020). Table 2 lists the IRE1/PERK inhibitors or activators, mentioning their known effect on mitochondrial function wherever applicable.

Table 2
www.frontiersin.org

Table 2. Inhibitors of IRE1 and PERK.

5 Discussion

Over the last decade, remarkable progress has been made towards understanding UPR signaling through two ER-resident kinases, IRE1 and PERK. While their role in ER stress-mediated signals is extensively explored, reports on their involvement in other physiological processes are still growing. Beyond proteotoxic stress, these regulations establish their non-canonical functions from development to diseases. IRE1 and PERK orchestrate diverse cellular processes, including protein folding, synaptic function, autophagy, calcium balance, and mitochondrial homeostasis via MAMs. Notably, dysregulated PERK/eIF2α and IRE1/XBP1 pathways are implicated in synaptic plasticity deficits, cognitive impairment, and maladaptive neuroinflammation. Conversely, these pathways are also essential at developmental stages during neurite outgrowth, underlining their dual role in development and disease progression. Similarly, these sensors maintain mitochondrial function and calcium flux at a normal physiological state. In contrast, during stress, these sensors help cells to adapt to quick changes in energy demands by modulating mitochondrial bioenergetics. Overall, due to these signaling overlaps, pharmacological interventions are challenging. A recent study by the Bertolotti group demonstrated concentration-dependent differential activation observed between nanomolar versus micromolar amounts. Interestingly, the authors demonstrated that ATP-competitive inhibitors of PERK, GSK2656157, GSK2606414, and AMG44, inhibit PERK in the nanomolar range but surprisingly activate the ISR via GCN2 at micromolar concentrations (Szaruga et al., 2023). Additionally, PERK inhibitors (GSK series) have been found to inhibit other kinases, causing additional challenges for their application as therapeutics (Rojas-Rivera et al., 2017). Additionally, due to its crucial role in high levels of insulin production in the pancreas, continuous administration of PERK inhibition leads to pancreatic toxicity (Yu et al., 2015). PERK inhibitor delayed Purkinje cell degeneration, reduced the skeletal muscle abnormalities with improved motor performance during the symptomatic phase in a mouse model of Marinesco-Sjögren syndrome (Grande et al., 2018). In contrast, PERK activators are beneficial against tauopathies and HD (Ganz et al., 2020; Li et al., 2025a). Thus, it is important to understand the cellular physiology and the pathological condition before administration of any small molecules modulating PERK’s function.

Similarly, limitations of IRE1α inhibitors is linked with off-target effects, target cell types, and the inherent dual expression nature of IRE1 (Siwecka et al., 2021). For example, IRE1 inhibitors have been applied in different cancer treatment. In multiple myeloma (MM), IRE1 has a tumor-growth-promoting role, while in B-cell lymphoma germinal center B-cell-like subtype (GCB-DLBCL), IRE1/XBP1s activity may negatively impact tumor growth (Bujisic et al., 2017). Thus, the same inhibition might have opposite effects based on the cancer types. The scenario changes in the context of brain aging, where the overall decline of the prototypical network has been observed with age. The genetic disruption of the ER stress sensor IRE1 accelerated age-related cognitive decline. Thus, a beneficial effect was observed after XBP1s gene delivery to aged mice. Similarly, boosting IRE1 ameliorates ALS disease phenotypes in C9orf72 mice by clearing poly (GR) dipeptide repeats (Li et al., 2025b).

Besides its large spectrum of activities IRE1, the action of IRE1 inhibitors also play an important role. STF-083010, an inhibitor of endonuclease activity of IRE1α, decreases maximal mitochondrial respiration and spare respiratory capacity in SH-SY5Y cells, and impairment of mitochondrial functions is independent of its inhibitory activity on IRE1α (Hatokova et al., 2023). Thus, a fine dosage depending on target cells is crucial for a beneficial effect. Moreover, a recent study has indicated IRE1 and PERK crosstalk during chronic ER stress, where IRE1 signaling increases PERK expression, suggesting involvement of crosstalk between two sensors (Ong et al., 2024). Considering these complex scenarios, dissecting these interrelations more carefully is necessary. An enhanced understanding of these two sensors is still required for better therapeutic interventions.

Author contributions

CK: Writing – original draft, Writing – review & editing. VK: Writing – original draft, Writing – review & editing. PK: Writing – original draft, Writing – review & editing. CG: Writing – original draft, Writing – review & editing. SM: Conceptualization, Data curation, Funding acquisition, Investigation, Project administration, Resources, Supervision, Visualization, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. SM acknowledges BITS-Pilani (Hyderabad Campus) for OPERA fellowship and the ICMR Ad-hoc research grant (project ID 2021-13896) for supporting research activities. CK and VK acknowledge BITS-Pilani (Hyderabad Campus) for institutional doctoral fellowship, and PK acknowledges CSIR/UGC/NFOBC for doctoral fellowship. CDG acknowledges fellowship from the ICMR Ad-hoc research grant (project ID 2021-13256).

Conflict of interest

The authors declare that the research 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) declare that no Gen AI was used in the creation of this manuscript.

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

Baron, K. R., Oviedo, S., Krasny, S., Zaman, M., Aldakhlallah, R., Bora, P., et al. (2025). Pharmacologic activation of integrated stress response kinases inhibits pathologic mitochondrial fragmentation. eLife 13:RP100541. doi: 10.7554/eLife.100541,

PubMed Abstract | Crossref Full Text | Google Scholar

Bassot, A., Chen, J., Takahashi-Yamashiro, K., Yap, M. C., Gibhardt, C. S., Le, G. N. T., et al. (2023). The endoplasmic reticulum kinase PERK interacts with the oxidoreductase ERO1 to metabolically adapt mitochondria. Cell Rep. 42:111899. doi: 10.1016/j.celrep.2022.111899,

PubMed Abstract | Crossref Full Text | Google Scholar

Behnke, J., Mann, M. J., Scruggs, F. L., Feige, M. J., and Hendershot, L. M. (2016). Members of the Hsp70 family recognize distinct types of sequences to execute ER quality control. Mol. Cell 63, 739–752. doi: 10.1016/j.molcel.2016.07.012,

PubMed Abstract | Crossref Full Text | Google Scholar

Bell, C., Bell, M. C., Meier, S. E., Ingram, A. L., and Abisambra, J. F. (2016). PERK-opathies: an endoplasmic reticulum stress mechanism underlying neurodegeneration. Curr. Alzheimer Res. 13, 150–163. doi: 10.2174/1567205013666151218145431,

PubMed Abstract | Crossref Full Text | Google Scholar

Bruch, J., Xu, H., Rösler, T. W., de Andrade, A., Kuhn, P. H., Lichtenthaler, S. F., et al. (2017). PERK activation mitigates tau pathology in vitro and in vivo. EMBO Mol. Med. 9, 371–384. doi: 10.15252/emmm.201606664,

PubMed Abstract | Crossref Full Text | Google Scholar

Bujisic, B., de Gassart, A., Tallant, R., Demaria, O., Zaffalon, L., Chelbi, S., et al. (2017). Impairment of both IRE1 expression and XBP1 activation is a hallmark of GCB DLBCL and contributes to tumor growth. Blood 129, 2420–2428. doi: 10.1182/blood-2016-09-741348,

PubMed Abstract | Crossref Full Text | Google Scholar

Cabral-Miranda, F., Tamburini, G., Martinez, G., Ardiles, A. O., Medinas, D. B., Gerakis, Y., et al. (2022). Unfolded protein response IRE1 / XBP1 signaling is required for healthy mammalian brain aging. EMBO J. 41:e111952. doi: 10.15252/embj.2022111952,

PubMed Abstract | Crossref Full Text | Google Scholar

Carreras-Sureda, A., Jaña, F., Urra, H., Durand, S., Mortenson, D. E., Sagredo, A., et al. (2019). Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat. Cell Biol. 21, 755–767. doi: 10.1038/s41556-019-0329-y,

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, K., Morizawa, Y. M., Nuriel, T., al-Dalahmah, O., Xie, Z., and Yang, G. (2025). Selective removal of astrocytic PERK protects against glymphatic impairment and decreases toxic aggregation of β-amyloid and tau. Neuron 113, 2438–2454.e6. doi: 10.1016/j.neuron.2025.04.027,

PubMed Abstract | Crossref Full Text | Google Scholar

Chiu, T.-L., Huang, H. Y., Chang, H. F., Wu, H. R., and Wang, M. J. (2023). Enhanced IRE1α phosphorylation/Oligomerization-triggered XBP1 splicing contributes to Parkin-mediated prevention of SH-SY5Y cell death under Nitrosative stress. Int. J. Mol. Sci. 24:2017. doi: 10.3390/ijms24032017,

PubMed Abstract | Crossref Full Text | Google Scholar

Chu, B., Li, M., Cao, X., Li, R., Jin, S., Yang, H., et al. (2021). IRE1α-XBP1 affects the mitochondrial function of Aβ25-35-treated SH-SY5Y cells by regulating mitochondria-associated endoplasmic reticulum membranes. Front. Cell. Neurosci. 15:614556. doi: 10.3389/fncel.2021.614556,

PubMed Abstract | Crossref Full Text | Google Scholar

Concha, N. O., Smallwood, A., Bonnette, W., Totoritis, R., Zhang, G., Federowicz, K., et al. (2015). Long-range inhibitor-induced conformational regulation of human IRE1α endoribonuclease activity. Mol. Pharmacol. 88, 1011–1023. doi: 10.1124/mol.115.100917,

PubMed Abstract | Crossref Full Text | Google Scholar

Corazzari, M., Gagliardi, M., Fimia, G. M., and Piacentini, M. (2017). Endoplasmic reticulum stress, unfolded protein response, and Cancer cell fate. Front. Oncol. 7:78. doi: 10.3389/fonc.2017.00078,

PubMed Abstract | Crossref Full Text | Google Scholar

Cross, B. C. S., Bond, P. J., Sadowski, P. G., Jha, B. K., Zak, J., Goodman, J. M., et al. (2012). The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proc. Natl. Acad. Sci. 109, E869–E878. doi: 10.1073/pnas.1115623109,

PubMed Abstract | Crossref Full Text | Google Scholar

Csordás, G., Renken, C., Várnai, 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

Daniele, J. R., et al. (2018). A non-canonical arm of UPRER mediates longevity through ER remodeling and lipophagy. Cell Biol. doi: 10.1101/471177

Crossref Full Text | Google Scholar

Duran-Aniotz, C., Cornejo, V. H., Espinoza, S., Ardiles, Á. O., Medinas, D. B., Salazar, C., et al. (2017). IRE1 signaling exacerbates Alzheimer’s disease pathogenesis. Acta Neuropathol. 134, 489–506. doi: 10.1007/s00401-017-1694-x,

PubMed Abstract | Crossref Full Text | Google Scholar

Edvardson, S., Nicolae, C. M., Noh, G. J., Burton, J. E., Punzi, G., Shaag, A., et al. (2019). Heterozygous RNF13 gain-of-function variants are associated with congenital microcephaly, epileptic encephalopathy, blindness, and failure to thrive. Am. J. Hum. Genet. 104, 179–185. doi: 10.1016/j.ajhg.2018.11.018,

PubMed Abstract | Crossref Full Text | Google Scholar

English, B. C., Savage, H. P., Mahan, S. P., Diaz-Ochoa, V. E., Young, B. M., Abuaita, B. H., et al. (2023). The IRE1α-XBP1 signaling Axis promotes glycolytic reprogramming in response to inflammatory stimuli. MBio 14:e0306822. doi: 10.1128/mbio.03068-22,

PubMed Abstract | Crossref Full Text | Google Scholar

Espina, M., di Franco, N., Brañas-Navarro, M., Navarro, I. R., Brito, V., Lopez-Molina, L., et al. (2023). The GRP78-PERK axis contributes to memory and synaptic impairments in Huntington’s disease R6/1 mice. Neurobiol. Dis. 184:106225. doi: 10.1016/j.nbd.2023.106225,

PubMed Abstract | Crossref Full Text | Google Scholar

Ganz, J., Shacham, T., Kramer, M., Shenkman, M., Eiger, H., Weinberg, N., et al. (2020). A novel specific PERK activator reduces toxicity and extends survival in Huntington’s disease models. Sci. Rep. 10:6875. doi: 10.1038/s41598-020-63899-4,

PubMed Abstract | Crossref Full Text | Google Scholar

Gerakis, Y., and Hetz, C. (2018). Emerging roles of ER stress in the etiology and pathogenesis of Alzheimer’s disease. FEBS J. 285, 995–1011. doi: 10.1111/febs.14332,

PubMed Abstract | Crossref Full Text | Google Scholar

Ghosh, R., Wang, L., Wang, E. S., Perera, B. G. K., Igbaria, A., Morita, S., et al. (2014). Allosteric inhibition of the IRE1α RNase preserves cell viability and function during endoplasmic reticulum stress. Cell 158, 534–548. doi: 10.1016/j.cell.2014.07.002,

PubMed Abstract | Crossref Full Text | Google Scholar

Grande, V., Ornaghi, F., Comerio, L., Restelli, E., Masone, A., Corbelli, A., et al. (2018). PERK inhibition delays neurodegeneration and improves motor function in a mouse model of Marinesco-Sjögren syndrome. Hum. Mol. Genet. 27, 2477–2489. doi: 10.1093/hmg/ddy152,

PubMed Abstract | Crossref Full Text | Google Scholar

Guirao-Abad, J. P., et al. (2020). A human IRE1 inhibitor blocks the unfolded protein response in the pathogenic fungus aspergillus fumigatus and suggests noncanonical functions within the pathway. mSphere 5, e00879–e00820. doi: 10.1128/mSphere.00879-20

Crossref Full Text | Google Scholar

Gundu, C., Arruri, V. K., Sherkhane, B., Khatri, D. K., and Singh, S. B. (2022). GSK2606414 attenuates PERK/p-eIF2α/ATF4/CHOP axis and augments mitochondrial function to mitigate high glucose induced neurotoxicity in N2A cells. Curr. Res. Pharmacol. Drug Disc. 3:100087. doi: 10.1016/j.crphar.2022.100087,

PubMed Abstract | Crossref Full Text | Google Scholar

Hatokova, Z., Evinova, A., and Racay, P. (2023). STF-083010 an inhibitor of IRE1α endonuclease activity affects mitochondrial respiration and generation of mitochondrial membrane potential. Toxicol. Vitro 92:105652. doi: 10.1016/j.tiv.2023.105652,

PubMed Abstract | Crossref Full Text | Google Scholar

Hayashi, T., and Su, T.-P. (2007). Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate ca(2+) signaling and cell survival. Cell 131, 596–610. doi: 10.1016/j.cell.2007.08.036,

PubMed Abstract | Crossref Full Text | Google Scholar

Hetz, C., Zhang, K., and Kaufman, R. J. (2020). Mechanisms, regulation and functions of the unfolded protein response. Nat. Rev. Mol. Cell Biol. 21, 421–438. doi: 10.1038/s41580-020-0250-z,

PubMed Abstract | Crossref Full Text | Google Scholar

Jacquemyn, J., Cascalho, A., and Goodchild, R. E. (2017). The ins and outs of endoplasmic reticulum-controlled lipid biosynthesis. EMBO Rep. 18, 1905–1921. doi: 10.15252/embr.201643426,

PubMed Abstract | Crossref Full Text | Google Scholar

Janikiewicz, J., Szymański, J., Malinska, D., Patalas-Krawczyk, P., Michalska, B., Duszyński, J., et al. (2018). Mitochondria-associated membranes in aging and senescence: structure, function, and dynamics. Cell Death Dis. 9:332. doi: 10.1038/s41419-017-0105-5,

PubMed Abstract | Crossref Full Text | Google Scholar

Kato, H., Okabe, K., Miyake, M., Hattori, K., Fukaya, T., Tanimoto, K., et al. (2020). ER-resident sensor PERK is essential for mitochondrial thermogenesis in brown adipose tissue. Life Sci. Alliance 3:e201900576. doi: 10.26508/lsa.201900576,

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, J., Jung, S. H., Kim, M., Park, W., Byun, J., Park, S., et al. (2024). Abstract 3335: A novel and potent IRE1α RNase inhibitor, HM100168 as a promising therapeutic strategy in solid cancers. Cancer Res. 84(6 _ Supplement):3335. doi: 10.1158/1538-7445.AM2024-3335,

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, S., Kim, D. K., Jeong, S., and Lee, J. (2022). The common cellular events in the neurodegenerative diseases and the associated role of endoplasmic reticulum stress. Int. J. Mol. Sci. 23:5894. doi: 10.3390/ijms23115894,

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, P., Scott, M. R., and Meador-Woodruff, J. H. (2021). Dysregulation of the unfolded protein response (UPR) in the dorsolateral prefrontal cortex in elderly patients with schizophrenia. Mol. Psychiatry 26, 1321–1331. doi: 10.1038/s41380-019-0537-7,

PubMed Abstract | Crossref Full Text | Google Scholar

Korennykh, A. V., Egea, P. F., Korostelev, A. A., Finer-Moore, J., Stroud, R. M., Zhang, C., et al. (2011). Cofactor-mediated conformational control in the bifunctional kinase/RNase Ire1. BMC Biol. 9:48. doi: 10.1186/1741-7007-9-48,

PubMed Abstract | Crossref Full Text | Google Scholar

Laguesse, S., Creppe, C., Nedialkova, D. D., Prévot, P. P., Borgs, L., Huysseune, S., et al. (2015). A dynamic unfolded protein response contributes to the control of cortical neurogenesis. Dev. Cell 35, 553–567. doi: 10.1016/j.devcel.2015.11.005,

PubMed Abstract | Crossref Full Text | Google Scholar

Latorre-Muro, P., O’Malley, K. E., Bennett, C. F., Perry, E. A., Balsa, E., Tavares, C. D. J., et al. (2021). A cold-stress-inducible PERK/OGT axis controls TOM70-assisted mitochondrial protein import and cristae formation. Cell Metab. 33, 598–614.e7. doi: 10.1016/j.cmet.2021.01.013,

PubMed Abstract | Crossref Full Text | Google Scholar

Lebeau, J., Saunders, J. M., Moraes, V. W. R., Madhavan, A., Madrazo, N., Anthony, M. C., et al. (2018). The PERK arm of the unfolded protein response regulates mitochondrial morphology during acute endoplasmic reticulum stress. Cell Rep. 22, 2827–2836. doi: 10.1016/j.celrep.2018.02.055,

PubMed Abstract | Crossref Full Text | Google Scholar

Li, Y., Liu, D., and Li, S. (2025b). IRE1/Xbp1 promotes the clearance of poly(GR) dipeptide repeats in amyotrophic lateral sclerosis. J. Biol. Chem. 301:110764. doi: 10.1016/j.jbc.2025.110764,

PubMed Abstract | Crossref Full Text | Google Scholar

Li, X., Lu, D., Zou, L., Ma, L., Yang, Y., Quan, X., et al. (2025a). Activation of the PERK branch of the UPR as a strategy for improving outcomes in acute ischemic stroke. ACS Omega 10, 15461–15470. doi: 10.1021/acsomega.5c00125,

PubMed Abstract | Crossref Full Text | Google Scholar

Lu, M., Lawrence, D. A., Marsters, S., Acosta-Alvear, D., Kimmig, P., Mendez, A. S., et al. (2014). Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science 345, 98–101. doi: 10.1126/science.1254312,

PubMed Abstract | Crossref Full Text | Google Scholar

Maity, S., Basak, T., Bhat, A., Bhasin, N., Ghosh, A., Chakraborty, K., et al. (2014). Cross-compartment proteostasis regulation during redox imbalance induced ER stress. Proteomics 14, 1724–1736. doi: 10.1002/pmic.201300449,

PubMed Abstract | Crossref Full Text | Google Scholar

Mimura, N., Fulciniti, M., Gorgun, G., Tai, Y. T., Cirstea, D., Santo, L., et al. (2012). Blockade of XBP1 splicing by inhibition of IRE1α is a promising therapeutic option in multiple myeloma. Blood 119, 5772–5781. doi: 10.1182/blood-2011-07-366633,

PubMed Abstract | Crossref Full Text | Google Scholar

Missiroli, S., Patergnani, S., Caroccia, N., Pedriali, G., Perrone, M., Previati, M., et al. (2018). Mitochondria-associated membranes (MAMs) and inflammation. Cell Death Dis. 9:329. doi: 10.1038/s41419-017-0027-2,

PubMed Abstract | Crossref Full Text | Google Scholar

Moreno, J. A., Halliday, M., Molloy, C., Radford, H., Verity, N., Axten, J. M., et al. (2013). Oral treatment targeting the unfolded protein response prevents neurodegeneration and clinical disease in prion-infected mice. Sci. Transl. Med. 5:206ra138. doi: 10.1126/scitranslmed.3006767,

PubMed Abstract | Crossref Full Text | Google Scholar

Mori, T., Hayashi, T., Hayashi, E., and Su, T. P. (2013). Sigma-1 receptor chaperone at the ER-mitochondrion interface mediates the mitochondrion-ER-nucleus signaling for cellular survival. PLoS One 8:e76941. doi: 10.1371/journal.pone.0076941,

PubMed Abstract | Crossref Full Text | Google Scholar

Muñoz, J. P., Ivanova, S., Sánchez-Wandelmer, J., Martínez-Cristóbal, P., Noguera, E., Sancho, A., et al. (2013). Mfn2 modulates the UPR and mitochondrial function via repression of PERK. EMBO J. 32, 2348–2361. doi: 10.1038/emboj.2013.168,

PubMed Abstract | Crossref Full Text | Google Scholar

Navarro-Betancourt, J. R., Papillon, J., Guillemette, J., Iwawaki, T., Chung, C. F., and Cybulsky, A. V. (2020). Role of IRE1α in podocyte proteostasis and mitochondrial health. Cell Death Discovery 6:128. doi: 10.1038/s41420-020-00361-4,

PubMed Abstract | Crossref Full Text | Google Scholar

Nevell, L., Zhang, K., Aiello, A. E., Koenen, K., Galea, S., Soliven, R., et al. (2014). Elevated systemic expression of ER stress related genes is associated with stress-related mental disorders in the Detroit neighborhood health study. Psychoneuroendocrinology 43, 62–70. doi: 10.1016/j.psyneuen.2014.01.013,

PubMed Abstract | Crossref Full Text | Google Scholar

Nishitoh, H., Matsuzawa, A., Tobiume, K., Saegusa, K., Takeda, K., Inoue, K., et al. (2002). ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 16, 1345–1355. doi: 10.1101/gad.992302,

PubMed Abstract | Crossref Full Text | Google Scholar

Ong, G., Ragetli, R., Mnich, K., Doble, B. W., Kammouni, W., and Logue, S. E. (2024). IRE1 signaling increases PERK expression during chronic ER stress. Cell Death Dis. 15:276. doi: 10.1038/s41419-024-06663-0,

PubMed Abstract | Crossref Full Text | Google Scholar

Papandreou, I., Denko, N. C., Olson, M., van Melckebeke, H., Lust, S., Tam, A., et al. (2011). Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma. Blood 117, 1311–1314. doi: 10.1182/blood-2010-08-303099,

PubMed Abstract | Crossref Full Text | Google Scholar

Park, G., Xu, K., Chea, L., Kim, K., Safarta, L., Song, K. H., et al. (2023). Neurodegeneration risk factor, EIF2AK3 (PERK), influences tau protein aggregation. J. Biol. Chem. 299:102821. doi: 10.1016/j.jbc.2022.102821,

PubMed Abstract | Crossref Full Text | Google Scholar

Perea, V., Cole, C., Lebeau, J., Dolina, V., Baron, K. R., Madhavan, A., et al. (2023). PERK signaling promotes mitochondrial elongation by remodeling membrane phosphatidic acid. EMBO J. 42:e113908. doi: 10.15252/embj.2023113908,

PubMed Abstract | Crossref Full Text | Google Scholar

Rahhali, K., Di Lisa, F., and Kaludercic, N. (2023). IP3 receptor trafficking at the ER-mitochondria contacts impacts on mitochondrial Ca2+ homeostasis and metabolism. Cell Calcium 110:102700. doi: 10.1016/j.ceca.2023.102700,

PubMed Abstract | Crossref Full Text | Google Scholar

Rendleman, J., Cheng, Z., Maity, S., Kastelic, N., Munschauer, M., Allgoewer, K., et al. (2018). New insights into the cellular temporal response to proteostatic stress. eLife 7:e39054. doi: 10.7554/eLife.39054,

PubMed Abstract | Crossref Full Text | Google Scholar

Ri, M., Tashiro, E., Oikawa, D., Shinjo, S., Tokuda, M., Yokouchi, Y., et al. (2012). Identification of Toyocamycin, an agent cytotoxic for multiple myeloma cells, as a potent inhibitor of ER stress-induced XBP1 mRNA splicing. Blood Cancer J. 2, e79. doi: 10.1038/bcj.2012.26,

PubMed Abstract | Crossref Full Text | Google Scholar

Rojas-Rivera, D., Delvaeye, T., Roelandt, R., Nerinckx, W., Augustyns, K., Vandenabeele, P., et al. (2017). When PERK inhibitors turn out to be new potent RIPK1 inhibitors: critical issues on the specificity and use of GSK2606414 and GSK2656157. Cell Death Differ. 24, 1100–1110. doi: 10.1038/cdd.2017.58,

PubMed Abstract | Crossref Full Text | Google Scholar

Sassano, M. L., Derua, R., Waelkens, E., Agostinis, P., and van Vliet, A. R. (2021). Interactome analysis of the ER stress sensor perk uncovers key components of ER-mitochondria contact sites and Ca2+ Signalling. Contact 4:25152564211052392. doi: 10.1177/25152564211052392,

PubMed Abstract | Crossref Full Text | Google Scholar

Sassano, M. L., van Vliet, A. R., Vervoort, E., van Eygen, S., van den Haute, C., Pavie, B., et al. (2023). PERK recruits E-Syt1 at ER–mitochondria contacts for mitochondrial lipid transport and respiration. J. Cell Biol. 222:e202206008. doi: 10.1083/jcb.202206008,

PubMed Abstract | Crossref Full Text | Google Scholar

Shacham, T., Patel, C., and Lederkremer, G. Z. (2021). PERK pathway and neurodegenerative disease: to inhibit or to activate? Biomolecules 11:354. doi: 10.3390/biom11030354,

PubMed Abstract | Crossref Full Text | Google Scholar

Shao, A., Xu, Q., Spalek, W. T., Cain, C. F., Kang, C. W., Tang, C. H. A., et al. (2020). Development of tumor-targeting IRE-1 inhibitors for B-cell Cancer therapy. Mol. Cancer Ther. 19, 2432–2444. doi: 10.1158/1535-7163.MCT-20-0127,

PubMed Abstract | Crossref Full Text | Google Scholar

Sharma, V., Ounallah-Saad, H., Chakraborty, D., Hleihil, M., Sood, R., Barrera, I., et al. (2018). Local inhibition of PERK enhances memory and reverses age-related deterioration of cognitive and neuronal properties. J. Neurosci. Off. J. Soc. Neurosci. 38, 648–658. doi: 10.1523/JNEUROSCI.0628-17.2017,

PubMed Abstract | Crossref Full Text | Google Scholar

Siwecka, N., Rozpędek-Kamińska, W., Wawrzynkiewicz, A., Pytel, D., Diehl, J. A., and Majsterek, I. (2021). The structure, activation and signaling of IRE1 and its role in determining cell fate. Biomedicine 9:156. doi: 10.3390/biomedicines9020156,

PubMed Abstract | Crossref Full Text | Google Scholar

Smith, H. L., and Mallucci, G. R. (2016). The unfolded protein response: mechanisms and therapy of neurodegeneration. Brain 139, 2113–2121. doi: 10.1093/brain/aww101,

PubMed Abstract | Crossref Full Text | Google Scholar

Son, S. M., Byun, J., Roh, S. E., Kim, S. J., and Mook-Jung, I. (2014). Reduced IRE1α mediates apoptotic cell death by disrupting calcium homeostasis via the InsP3 receptor. Cell Death Dis. 5:e1188. doi: 10.1038/cddis.2014.129,

PubMed Abstract | Crossref Full Text | Google Scholar

Szaruga, M., Janssen, D. A., de Miguel, C., Hodgson, G., Fatalska, A., Pitera, A. P., et al. (2023). Activation of the integrated stress response by inhibitors of its kinases. Nat. Commun. 14:5535. doi: 10.1038/s41467-023-40823-8,

PubMed Abstract | Crossref Full Text | Google Scholar

Takeda, K., Nagashima, S., Shiiba, I., Uda, A., Tokuyama, T., Ito, N., et al. (2019). MITOL prevents ER stress-induced apoptosis by IRE1α ubiquitylation at ER-mitochondria contact sites. EMBO J. 38:e100999. doi: 10.15252/embj.2018100999,

PubMed Abstract | Crossref Full Text | Google Scholar

Tam, A. B., Koong, A. C., and Niwa, M. (2014). Ire1 has distinct catalytic mechanisms for XBP1/HAC1 splicing and RIDD. Cell Rep. 9, 850–858. doi: 10.1016/j.celrep.2014.09.016,

PubMed Abstract | Crossref Full Text | Google Scholar

Urano, F., Wang, X., Bertolotti, A., Zhang, Y., Chung, P., Harding, H. P., et al. (2000). Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science (New York, N.Y.) 287, 664–666. doi: 10.1126/science.287.5453.664,

PubMed Abstract | Crossref Full Text | Google Scholar

Vance, J. E. (1990). Phospholipid synthesis in a membrane fraction associated with mitochondria. J. Biol. Chem. 265, 7248–7256. doi: 10.1016/S0021-9258(19)39106-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Verfaillie, T., Rubio, N., Garg, A. D., Bultynck, G., Rizzuto, R., Decuypere, J. P., et al. (2012). PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 19, 1880–1891. doi: 10.1038/cdd.2012.74,

PubMed Abstract | Crossref Full Text | Google Scholar

Wan, H., Wang, Q., Chen, X., Zeng, Q., Shao, Y., Fang, H., et al. (2020). WDR45 contributes to neurodegeneration through regulation of ER homeostasis and neuronal death. Autophagy 16, 531–547. doi: 10.1080/15548627.2019.1630224,

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, X., Zhu, X., Huang, G., Wu, L., Meng, Z., and Wu, Y. (2023). Knockout of PERK protects rat Müller glial cells against OGD-induced endoplasmic reticulum stress-related apoptosis. BMC Ophthalmol. 23:286. doi: 10.1186/s12886-023-03022-z,

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, S., Xu, Y., Chen, L., Fang, Q., Song, S., Chen, J., et al. (2017). RCN1 suppresses ER stress-induced apoptosis via calcium homeostasis and PERK-CHOP signaling. Oncogene 6:e304. doi: 10.1038/oncsis.2017.6,

PubMed Abstract | Crossref Full Text | Google Scholar

Xue, X., Wu, X., Liu, L., Liu, L., and Zhu, F. (2023). ERVW-1 activates ATF6-mediated unfolded protein response by decreasing GANAB in recent-onset schizophrenia. Viruses 15:1298. doi: 10.3390/v15061298,

PubMed Abstract | Crossref Full Text | Google Scholar

Yan, F., Cao, S., Li, J., Dixon, B., Yu, X., Chen, J., et al. (2017). Pharmacological inhibition of PERK attenuates early brain injury after subarachnoid hemorrhage in rats through the activation of Akt. Mol. Neurobiol. 54, 1808–1817. doi: 10.1007/s12035-016-9790-9,

PubMed Abstract | Crossref Full Text | Google Scholar

Yan, C., Liu, J., Gao, J., Sun, Y., Zhang, L., Song, H., et al. (2019). IRE1 promotes neurodegeneration through autophagy-dependent neuron death in the Drosophila model of Parkinson’s disease. Cell Death Dis. 10:800. doi: 10.1038/s41419-019-2039-6,

PubMed Abstract | Crossref Full Text | Google Scholar

Yu, Q., Zhao, B., Gui, J., Katlinski, K. V., Brice, A., Gao, Y., et al. (2015). Type I interferons mediate pancreatic toxicities of PERK inhibition. Proc. Natl. Acad. Sci. 112, 15420–15425. doi: 10.1073/pnas.1516362112,

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, K., Huang, Q., Li, X., Zhao, Z., Hong, C., Sun, Z., et al. (2024). The cGAS-STING pathway in viral infections: a promising link between inflammation, oxidative stress and autophagy. Front. Immunol. 15:1352479. doi: 10.3389/fimmu.2024.1352479,

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, D., Liu, Y., Zhu, Y., Zhang, Q., Guan, H., Liu, S., et al. (2022). A non-canonical cGAS-STING-PERK pathway facilitates the translational program critical for senescence and organ fibrosis. Nat. Cell Biol. 24, 766–782. doi: 10.1038/s41556-022-00894-z,

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, Q., Che, X., Zhang, H., Fan, P., Tan, G., Liu, L., et al. (2017). Thioredoxin-interacting protein links endoplasmic reticulum stress to inflammatory brain injury and apoptosis after subarachnoid haemorrhage. J. Neuroinflammation 14:104. doi: 10.1186/s12974-017-0878-6,

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: ER-mitochondrial interactions, neurodegenerative diseases, ER stress sensors, mitochondrial health, IRE1, pERK, UPR signaling

Citation: Kunja C, Kumar V, Kodam P, Gopu CD and Maity S (2025) ER stress sensors at the ER-mitochondrial interface, controlling mitochondrial health in neurodegenerative diseases. Front. Neurosci. 19:1665272. doi: 10.3389/fnins.2025.1665272

Received: 13 July 2025; Accepted: 30 October 2025;
Published: 15 December 2025.

Edited by:

Youngshin Lim, Cedars Sinai Medical Center, United States

Reviewed by:

Richard M. Monaghan, The University of Manchester, United Kingdom
Margret Bülow, University of Bonn, Germany

Copyright © 2025 Kunja, Kumar, Kodam, Gopu and Maity. 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: Shuvadeep Maity, c2h1dmFkZWVwQGh5ZGVyYWJhZC5iaXRzLXBpbGFuaS5hYy5pbg==

These authors share first authorship

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.