- Review
- Open access
- Published:
The integrated stress response in neurodegenerative diseases
Molecular Neurodegeneration volume 20, Article number: 20 (2025)
Abstract
The integrated stress response (ISR) is a conserved network in eukaryotic cells that mediates adaptive responses to diverse stressors. The ISR pathway ensures cell survival and homeostasis by regulating protein synthesis in response to internal or external stresses. In recent years, the ISR has emerged as an important regulator of the central nervous system (CNS) development, homeostasis and pathology. Dysregulation of ISR signaling has been linked to several neurodegenerative diseases. Intriguingly, while acute ISR provide neuroprotection through the activation of cell survival mechanisms, prolonged ISR can promote neurodegeneration through protein misfolding, oxidative stress, and mitochondrial dysfunction. Understanding the molecular mechanisms and dynamics of the ISR in neurodegenerative diseases aids in the development of effective therapies. Here, we will provide a timely review on the cellular and molecular mechanisms of the ISR in neurodegenerative diseases. We will highlight the current knowledge on the dual role that ISR plays as a protective or disease worsening pathway and will discuss recent advances on the therapeutic approaches that have been developed to target ISR activity in neurodegenerative diseases.
Introduction
The integrated stress response (ISR) is an evolutionarily conserved mechanism to cellular stress that is activated by different intrinsic and extrinsic factors. The main function of the ISR is to maintain cellular homeostasis by downregulating protein synthesis and upregulating specific target genes [1]. The key intrinsic stress is endoplasmic reticulum stress (ER stress), which occurs when the capacity of protein folding is exceeded [1, 2]. Extrinsic stressors include glucose and amino acid deprivation, hypoxia, viral infections, and the presence of reactive oxygen species (ROS) [1]. The ISR works with several other cellular adaptation pathways such as proteotoxicity, ubiquitin–proteasome, autophagy, phosphatidylinositol‐3 kinase (PI3K) signaling, and unfolded protein response (UPR); which act in a time-dependent manner upon induction of any stress mediated signaling [3,4,5,6,7,8,9,10]. Moreover, the cross-talk of ISR with osmotic stress response (OSR), DNA damage response (DDR), and heat-shock protein (HSP) response is reported and is often known to function in a cytoprotective manner [11,12,13,14].
There are four regulatory ISR-kinases, all of which converge on the phosphorylation of the alpha subunit of the eukaryotic initiator factor 2 (eIF2α) at the serine site 51 [15]. Phosphorylation of eIF2α reduces global protein synthesis and the formation of stress granules are also reported in some cases, while promoting the translation of stress-related genes such as activating transcription factor 4 (ATF4) [16], which supports cell survival. However, under conditions of severe cellular stress, the adaptive response loses its ability to effectively alleviate the stress and instead promotes the activation of apoptotic pathways [17, 18]. The ISR is terminated with the dephosphorylation of eIF2α in a negative feedback loop manner. The ISR has been observed to be activated in different neurodegenerative disorders like multiple sclerosis (MS), Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS) [19]. This review outlines ISR mechanisms, and its relevance in CNS homeostasis and in neurodegenerative disorders where ER stress and ISR link together to contribute to neuronal cell death, inflammation, protein aggregation, and cognitive impairment. We will also provide an overview on the current therapeutic strategies targeted toward the modulation of dysregulated ISR in neurodegenerative disorders.
Initiation of the ISR: Four sensors, one phosphorylation
The ISR is first initiated by a disturbance in homeostasis and is recognized by the eIF2α protein kinases (EIF2AKs). When changes in homeostasis are detected, the kinases phosphorylate the alpha subunit of the eukaryotic initiator factor 2 (eIF2α) to inhibit protein synthesis, reconfigure gene expression for stress adaptation or inducing apoptosis [1]. The four kinases are PKR-like ER kinase (PERK), protein kinase double-stranded RNA dependent (PKR), general control non-derepressible-2 (GCN2), and heme-regulated inhibitor (HRI) [15, 20]. While each kinase has its own regulatory mechanisms, they all converge on the phosphorylation of eIF2α at the serine site 51 (Fig. 1), which is critical for translation control [21, 22]. PERK, also known as EIF2AK3, is an ER transmembrane protein that becomes activated upon detecting disturbances in the ER, like the accumulation of misfolded or unfolded proteins, calcium depletion, or redox imbalance [1, 23, 24]. There are two proposed models for PERK activation. One model suggests that, in homeostatic conditions, PERK remains inactive as its luminal domain is bound to GRP78. Upon ER stress, GRP78 dissociates from PERK to bind with unfolded proteins, leading to PERK dimerization and autophosphorylation that initiates downstream signaling [9, 25, 26]. The second and most recent model suggests an alternative mechanism in which PERK can be activated directly by binding to unfolded or misfolded proteins via its luminal domain [27, 28]. Once activated, PERK then phosphorylates eIF2α (p-eIF2α), preventing the translation of mRNA and reducing protein synthesis, which allows the ER to either refold or dispose of the misfolded proteins [15]. Simultaneously, p-eIF2α initiates the translation of ISR-specific mRNAs, like ATF4 [1].
The integrated stress response signaling pathway. The ISR can be initiated upon sensing ER stress, dsRNA, amino acid deprivation, and heme depletion, which are recognized by PERK, PKR, GCN2, and HRI respectively. Once activated, the four kinases converge in the phosphorylation of eIF2α that then activates ATF4. ATF4 translocate to the nucleus in which promotes the translation of stress response genes. Evidence shows regulation of ISR through “negative feedback”. ATF4 induces the expression of GADD34, which promotes the dephosphorylation of eIF2α by recruiting CReP and PP1c, hence restoring protein synthesis. Figure was created in BioRender
Unfolded proteins are not the only trigger for PERK autophosphorylation. It has been reported that saturation of lipids and modifications in the lipid composition of the ER membrane can trigger PERK activation [29]. Moreover, early studies reported that glucose deprivation in cultured hippocampal neurons leads to PERK activation and an increased expression of caspase-12, an ER resident caspase that has been associated with stress-induced apoptosis [30]. PKR (EIF2AK2) is found in the cytosol and nucleus of mammalian cells [1, 15, 31]. In addition to mounting an interferon response to viral infection [3], PKR also responds to other stimuli such as metabolic and ER stress [31, 32], as well as oxidative stress [33]. Upon stress, PKR dimerizes and auto-phosphorylates, forming a PKR-PKR complex that phosphorylates eIF2α [15]. Studies have shown that prolonged activation of PKR after oxidative stress increased the sensitivity to apoptosis, making its downregulation important for promoting cell survival [33, 34]. Importantly, PKR induces activation of the pro-apoptotic factor C/EBP homologous protein, CHOP, in response to hyperoxia [35]. GCN2 (EIF2AK4) is proposed to regulate changes in gene expression due to acid and glucose deprivation by sensing uncharged transfer ribonucleic acid (tRNA), although the exact mechanism is yet to be elucidated [15, 36]. Under amino acid deprivation, tRNA accumulates in the A site of the ribosome where it is recognized by the GCN2 regulatory domain histidyl-tRNA synthetase (HisRS), triggering GCN2 dimerization, eIF2α phosphorylation, and ISR activation [4]. GCN2 can also sense other stresses. For instance, inhibiting the proteasome system leads to the formation of stress granules that are primarily recognized by GCN2 and initiates the ISR [37, 38]. HRI or EIF2AK1 is mainly found in erythroid cells. Early studies reported that HRI is involved in erythrocyte differentiation and is required to produce α and β globin in red blood cell (RBC) precursors and it also promotes the survival of these cells under heme deficiency [39]. When the availability of iron decreases, eIF2α phosphorylation by HRI inhibits the translation of globin mRNAs, prevents hemoglobin production and exerts protection against toxic globin aggregates [1, 39]. HRI has also been found to initiate autophagy in cases when α-synuclein is overexpressed, hence helping to clear out the protein aggregates accumulated in the cytosol [40]. It is also demonstrated that HRI is required for inflammatory responses during infection [41]. Recent studies show that HRI acts as a proteotoxicity sensor via a pathway involving Hsp70, Bag3 and HRI, which detects the abnormal accumulation of proteins in the cytosol and triggers the phosphorylation of eIF2α [42]. All four kinases phosphorylate eIF2α at its serine site 51 and with this in common, they can often overlap and act cooperatively to sense different stress stimuli and integrate them to achieve specific cell responses, hence the name “integrated stress response”.
Downstream p-eIF2a, cellular responses, and termination of the ISR
Under homeostatic conditions, eIF2 participates in mRNA translation and recognition of the initiation codon AUG [43]. eIF2 is a 126 kDa heterotrimer protein comprised of α, β and γ subunits [44] with the eIF2α subunit playing a major regulatory role due to its RNA binding and phosphorylation sites [1]. Translation initiation involves the assembling of elongation-competent 80S ribosomes with an initiator tRNA at the ribosomal P site 1. This two-step process requires at least 9 eukaryotic initiation factors including eIF2. The first step requires the formation of 48S initiation complexes, which then join 60S subunits [43]. Many of the 48S initiation complexes are formed by a 43S preinitiation complex which is comprised of a 40S subunit, the eIF2-GTP-Met-tRNAiMet ternary complex, and other eIFs like eIF3, eIF1, eIF1A, and eIF5. This complex is composed of eIF2, guanosine triphosphate (GTP), and charged methionyl-transfer RNA (Met-tRNAiMet) [43, 45]. Upon stress, p-eIF2α alters the regular translation initiation by inhibiting the formation of active GTP from the eIF2-GDP complex and binds strongly to a modulatory portion of eIF2β that inhibits the formation of active GTP from the eIF2-GDP bound form. Consequently, there is less availability of the ternary complex, which leads to decreased translation rates (Fig. 1) and increased translation of ISR-related mRNAs like ATF4, ATF5, CHOP, and GADD34 [1, 45].
ATF4 is a key regulator in the ISR network, as it is vital for relieving ER stress by either promoting adaptation or triggering apoptosis [1]. ATF4 downstream activity initiates with the formation of homo and or heterodimers with other basic leucine zipper (bZIP) transcription factors including CHOP or AP-1 members that are known to regulate the transcriptional selectivity and thereby influence the outcome of ISR [46, 47]. The interacting heterodimers bind to cAMP (cyclic adenosine monophosphate) responsive elements to control target gene expression [18], which leads to the transcriptional upregulation of stress-related genes and pathways related to amino acid transport and metabolism as ATF4 can promote cell survival by inducing autophagy. Following amino acid starvation and ER stress, GCN2 and PERK along with ATF4 and CHOP can promote the expression of genes related to the autophagosome formation and function [48]. Dimer combinations between ATF4 and CHOP can regulate transcription through various mechanisms. In response to leucine starvation, ATF4-CHOP heterodimers regulate genes related to the degradation of ubiquitinated substrates, such as Nbr1, Atg7, and p62 [48]. Autophagy-related genes Atg10, Gabarap, and Atg5 are expressed after the formation of ATF4-CHOP heterodimer in response to amino acid deprivation [48]. Other reports suggest that ATF4 alone can target genes related to amino acid transport and biosynthesis, and it can act along with CHOP to regulate several shared genes related to protein synthesis, mRNA translation, and the unfolded protein response (UPR) [17]. The UPR is a signaling pathway that becomes activated due to ER stress and detects unfolded proteins through ER transmembrane receptors: PERK, IRE1, and ATF6. Both the ISR and UPR converge in the phosphorylation of eIF2α through PERK (Fig. 2) [1]. eIF2α phosphorylation can either promote cell survival or cell death. The increase in protein synthesis mediated by ATF4 and CHOP can promote cell death by oxidative stress and depletion of ATP [1, 18, 49]. This dual role of ATF4 has been attributed to the formation of heterodimers with different binding partners that lead to different responses. Binding partners of ATF4, such as C/EBPβ and C/EBPγ are involved in signal adaptation, upregulation of stress response genes, and protection against oxidation [50, 51]; while ATF4-CHOP is mainly associated with autophagy and pro-apoptotic activity. As reviewed previously by others [52], CHOP has been identified to induce apoptosis; however, as described earlier, it can also promote cell survival. This discrepancy may reflect the duration and level of stress, as well as the level of CHOP expression. These findings suggest that ATF4/CHOP may promote an initial survival response; however, with prolonged stress CHOP expression will initiate cell death to restore homeostasis [52].
Mitochondrial dysfunction and ISR: The activation of the integrated stress response (ISR) during mitochondrial dysfunction is triggered by various mechanisms. The fragmentation of mitochondrial DNA is among the leading causes of mitochondrial dysfunction, which is managed by an ISR sensor, protein kinase RNA-activated (PKR). Mitochondrial dysfunction during amino acid metabolism is managed by another class of ISR kinase- general control nonderepressible-2 (GCN2). The Tri-Carboxylic Acid (TCA) cycle is fed via the amino acid degradation during metabolic rewiring stage of mitochondrial stress, and the depletion of amino acids results in activation of GCN2. During this process, the equilibrium maintenance of reducing equivalents is maintained by the malate and aspartate shuttle. Upon induction of mitochondrial stress by generation of reactive oxygen species (ROS), a mitochondrial protease known as OMA1 regulates the mitochondrial stress dynamics by cleaving DAP3-binding cell death enhancer 1 (DELE1). This subsequently activates heme-regulated inhibitor (HRI) after translocating to the cytoplasm. The crosstalk between mitochondria and endoplasmic reticulum aids ER to sense the alterations in the levels of calcium (Ca2 +), ROS, and changes in energy productions; leading to activation of ER transmembrane protein- PKR-like ER kinase (PERK). Upon mitochondrial dysfunction and the ER-mitochondrial crosstalk, these protein kinases phosphorylate eIF2α and inhibit global protein translation to mitigate stress and restore the normal homeostasis. Until the stress is resolved, a translational shift due to upstream open reading frame (uORF) mediates selective translation of proteins such as ATF-4 and usurps the global translation to mitigate the stress. ATF-4 along with its other dimerization partners regulates amino acid synthesis genes, antioxidant pathways, chaperones and metabolism related genes to restore the homeostasis post stress. However, chronic stress results in activation of CHOP-mediated apoptotic cell death. Figure was created in BioRender
The termination of the ISR, restoration of protein synthesis, and return to homeostasis occur upon eIF2α dephosphorylation in a process regulated by the protein phosphatase 1 (PP1) complex through its catalytic subunit (PP1c). The phosphatase activity can be regulated by GADD34 (growth arrest and DNA damage-inducible protein 34) or by the “constitutive repressor of eIF2α phosphorylation”, known as CReP [53, 54]. CReP forms a complex with PP1c to enable homeostasis by maintaining low activity of p-eIF2α, whereas GADD34 expression is induced during the late stages of the ISR in response to ATF4 and forms a complex with PP1 to promote eIF2α dephosphorylation. Thus, this negative feedback loop appears to be essential for restoring homeostasis after a stress response [1, 53].
ISR and mitochondrial UPR
Mitochondrial stress responses (MSRs) can also result in the buildup of misfolded or damaged proteins. Mutations or deletions in mitochondrial DNA (mtDNA) causes an accumulation of unfolded proteins, which can trigger the activation of the mitochondrial unfolded protein response (UPRmt) [55]. Growing evidence has implicated ISR in UPRmt [56, 57] (Fig. 2). Studies in S. cerevisiae and C. elegans indicate that UPRmt is regulated by ISR sensors or eIF2α kinases. GCN2 depletion is known to significantly upregulate the expression of mitochondrial chaperones that activates UPRmt [55]. Another study revealed that during mitochondrial stress, activation of HRI, even with the absence of full heme, attenuates the global protein translation through phosphorylation of eIF2α, thus implicating a cross-talk between ISR and mitochondrial dysfunction [58]. During the active state of ISR and upon diverse mitochondrial insults, the upstream open reading frames (uORF’s) are involved in translation of selective transcription factors such as ATF4, ATF5 and CHOP [59,60,61]. The accumulation of misfolded or unfolded proteins in the mammalian mitochondrial matrix often leads to the upregulation of chaperones in mitochondria but no ER stress protein response is initiated [61]. Interestingly, UPRmt activation also leads to a reduction in the transcription of oxidative phosphorylation (OXPHOS) components, as mitochondria attempt to lessen their functional demands while addressing the stress [62, 63]. The mitochondrial stress modulators such as FCCP, doxycycline, MitoBloCk and actinonin are known to activate ATF4 and suppress the mitochondrial translation, alleviating the burden of misfolded proteins [62, 64]. ATF5 is also known to have direct effects on UPRmt, as inhibiting its function suppresses the induction of UPRmt-related genes during the mitochondrial stress. However, ATF5 overexpression in C. elegans lacking ATFS-1 is reported to restore the expression of HSP60 [62, 64]. These findings indicate a dual functionality of ATF-5 (or its isoform ATFS-1 in C. elegans) in regulation of mitochondrial dysfunction that needs further examination. Altogether, mitochondrial dysfunction and its association with other organelles that initiate stress response cascade have been reported in various disease conditions including neurodegenerative disorders [65,66,67], which is further discussed in detail in neurodegenerative disease section.
Pharmacological modulation of the ISR
The ISR has been associated with inflammation, cancer, diabetes, and neurodegenerative disorders. Hence, the ISR has become a promising therapeutic target [45]. Due to the dual roles that the ISR plays in promoting cell survival or cell death, different strategies have been developed to either enhance or suppress the ISR by targeting eIF2α, eIF2α kinases, or ATF4 [1] (Table 1). Pharmacological enhancement of the ISR can be attained by direct targeting of eIF2α kinases or by prolonging the phosphorylation of eIF2α with phosphatase inhibitors [1]. A common ISR activator is CCT020312, a small molecule that selectively targets PERK and promotes the phosphorylation of eIF2α without eliciting a general UPR [68]. BTdCPU is an active N,N’-diarylurea that acts as a potent HRI activator [69]. Drugs like salubrinal, guanabenz, and Sephin1 are known to prolong the ISR, which is achieved by inhibiting eIF2α dephosphorylation [1]. Salubrinal is a phosphatase inhibitor that selectively hinders eIF2alpha dephosphorylation independent of the eIF2α kinases. Although its mechanism of action is yet to be elucidated, evidence suggests it seems to inhibit CReP-PP1 and GADD34-PP1 complexes [70]. Guanabenz, a drug initially used as a treatment for hypertension, is shown to increase eIF2α phosphorylation by inhibiting GADD34 [71]. However, Guanabenz does not selectively inhibit GADD34 and its affinity for the α2-adrenergic receptor can result in adverse effects like drowsiness and lethargy upon overdose [72]. Hence, Guanabenz derivatives have been investigated. Sephin1 is a promising Guanabenz derivative that selectively inactivates the binding of GADD34 to PP1c without causing α2-adrenergic-related side effects in vitro or in vivo [73]. Taken together, Salubrinal, Guanabenz, and Sephin1 prolong the ISR and appear to decrease protein synthesis and allow protein folding, thus they are currently the most promising candidates to mitigate proteotoxicity [1].
While the ISR can have cytoprotective effects, its chronic activation can result in neurological disorders. Hence, pharmacological inhibitors of the eIF2α kinases have pursued as therapeutic targets [1]. One of the most well-known ISR inhibitors is the integrated stress response inhibitor (ISRIB). First identified by Sidrauski and colleagues, ISRIB acts downstream of all ISR-kinases and reverses the outcomes of eIF2α phosphorylation and restoring translation [74]. Several studies have demonstrated that ISRIB binds to the eIF2B complex, a guanine nucleotide exchange factor (GEF) for eIF2, and it stabilizes it in a structure that can exchange GDP for GTP on eIF2, thus promoting the restoration of protein synthesis [75,76,77,78]. ISRIB has been demonstrated to inhibit the formation of stress granules due to eIF2α phosphorylation and is considered a promising candidate treatment for diseases associated with the formation of protein aggregates [74]. However, recent studies have shown that ISRIB can only suppress the ISR when the levels of p-eIF2α are below a critical threshold [79]. Additionally, by using an ATF4 reporter cell line, it was found that ISRIB could not hinder the expression of stress-induced proteins under high intracellular levels of p-eIF2α. Thus, ISRIB seems to be effective under conditions of limited stress, and further investigation is required to determine if this property would affect its efficacy when used as a treatment for ISR-related diseases [79]. Other eIF2α inhibitors include the ATP-competitive PERK inhibitor GSK2606414 which prevents PERK autophosphorylation [80, 81] or the small molecule C16 which blocks autophosphorylation of PKR [82]. A recent work has reported that the commonly used ATP-competitive PERK inhibitorGSK2606414, can effectively inhibit PERK when used in nanomolar concentrations; however, it could activate GCN2 when used in micromolar concentrations [83]. Similarly, the PKR inhibitor, C16, was found to activate GCN2. In contrast, the GCN2 inhibitor A92 was shown to be effective in silencing its target but ultimately induces the ISR by activating PERK [83]. Altogether, extensive progress has been made in modulating the ISR in different preclinical in vivo and in vitro models (Table 1). Nonetheless, the complexity and redundancy of the stress-sensing pathways of the ISR requires further investigation in preclinical studies to help refine the selectivity of activators and inhibitors in future clinical settings.
The ISR in CNS homeostasis
The ISR pathway is fundamental for maintaining CNS homeostasis during neurodevelopment and adulthood and play important roles in CNS pathology [84]. Developmental studies suggest the existence of a non-canonical eIF2α pathway that selectively modulates changes in protein synthesis in axons and is required for neural wiring [85]. As an example, Sempahorin-3A (Sema3A), an extrinsic cue that can initiate protein synthesis in axons, is shown to induce eIF2α phosphorylation by activating PERK [85]. However, activation of sema3A signaling does not result in the suppression of global protein translation, nor the translation of ATF4. ISR is also implicated in the onset of some neurodevelopmental disorders such as Fragile X syndrome (FXS) and Down’s syndrome [86]. These disease models are known to exhibit impaired synaptic plasticity, which are hypothesized to be central to the clinical symptomatology [86].
Homeostasis of the CNS and proper function of neural network rely on synaptic transport and communication, which itself requires turnover of large amounts of proteins [87]. Evidence shows that the ISR can be activated by an abnormal increase in protein synthesis [84]. Given memory formation requires continuous protein synthesis there has been a growing interest in unraveling whether ISR plays a role in memory. Current findings suggest that the ISR negatively regulates memory formation. Recent research underscores the critical role of translation, particularly the bidirectional function of eIF2α phosphorylation, in long-term memory potentiation (LTP) and synaptic plasticity [88]. Synaptic plasticity is an underlying process of experience-dependent synaptic strength remodeling that is crucial for learning and memory processes. A nuanced interaction of released neurotransmitters, variety and their number of postsynaptic receptors could potentially cause the overall consolidation or diminishment of synaptic connections [89, 90]. De-novo protein synthesis is often required for long lasting synaptic plasticity through long-term potentiation (LTP; strengthening of synapse) and long-term depression (LTD; weakening of synapse) [91]. The ISR is known to regulate these protein synthesis pathways, and its effects have been extensively studied in a variety of brain regions of a broad range of species of organisms [19, 92,93,94].
Studies in the mouse forebrain demonstrate that the specific inhibition of C/EBP-family proteins facilitate long-term plasticity and memory by decreasing ATF4 expression, enhancing hippocampal-dependent spatial memory, and decreasing LTPs threshold [95] GCN2 also contributes to regulating synaptic plasticity, learning, and memory in mice through the modulation of the ATF4/CREB pathway [87]. In contrast, a reduction in the phosphorylation of GCN2 and eIF2α is associated with an LTP-induced stimulus, which can enhance both synaptic plasticity and memory formation [87]. Short-term working memory is a temporary process that does not rely on protein synthesis [96]. It is, however, highly dependent on Ca2 + dynamics. Interestingly, ablation of PERK from the forebrain of adult mice impairs working memory. Although the underlying mechanisms are yet to be identified, it is speculated that PERK mediates working memory by modifying the Ca2 + dynamics [96]. ATF4 is also involved in neurodevelopment by regulating the post-synaptic development of dendritic spines that regulate neuronal activity and are important for synaptic plasticity and memory [97, 98]. Injection of shRNA targeting PERK into the CA1 region of the hippocampus of young adult mice is shown to decrease PERK expression in excitatory and inhibitory neurons, which results in enhanced neuronal excitability and improved cognitive function and hippocampal-dependent learning [99].
Astrocytic protein synthesis plays a major role in modulation of synaptic plasticity and long-term memory consolidation [35]. In a mouse model of genetically ablated Ser51 phosphorylation site of eIF2α (Eif2aA/A) in astrocytes, a reduced p-eIF2α expression resulted in increased protein synthesis which accounts for increased excitatory and decreased inhibitory synaptic outputs to the pyramidal neurons [100]. This highlights the role of autonomous and cell-type specific translational control in memory consolidation [35, 100, 101]. In addition to memory consolidation, astrocytes play a crucial role in maintaining CNS homeostasis by various mechanisms including metabolic support of neurons and maintaining the integrity of the blood brain barrier (BBB) [102]. Upon specific stimuli, including the activation of the ISR, astrocytes become reactive. Astrocyte reactivity in neurodegenerative diseases is often associated with disruption of BBB that promotes neuroinflammation and neurodegeneration [102]. Modulators of the ISR are shown to have cytoprotective effects in CNS injuries and diseases by restoring BBB. For example, studies in a mouse model of CNS stab injury showed that Salubrinal can restore the BBB by increasing the expression of fibronectin and reducing the activation of microglia and macrophages [103]. Likewise, in a rat model of cerebral ischemia, administration of Salubrinal attenuated the expression levels of metalloprotease 9 (MMP-9); a known marker of BBB impairment, as well as adhesion molecules and mediators of leukocyte migration factors, namely ICAM-1 and VCAM-1 [104]. These findings suggest a potential for Salubrinal, as an ISR modulator, in restoring BBB [104,105,106,107].
ATF-4 is also known to play an evolutionary conserved role in maintaining CNS metabolism and cellular redox capacity by regulating amino acid biosynthesis, cysteine in particular, with antioxidant effects [108, 109]. In Parkin and Pink mutants of Parkinsonian Drosophila models with a dysfunctional mitochondria-mediated oxidative stress, ATF4 rescues the loss of dopaminergic neurons [108, 109]. In a model of amino acid deprivation, GCN2 also supports amino acid metabolism by its direct interaction with deacetylated tRNAs through histidyl tRNA synthetase (HisRS-like) [110, 111]. A growing body of evidence from animal models of neurodegenerative diseases indicates that sustained phosphorylation of eIF2α is linked to cognitive deficits [112, 113]. The protective effects of GCN2-mediated ISR signaling in nmf205 mouse model of neurodegeneration offers a unique perspective on how the interaction of ribosome with GCN2 acts as a feedback loop during ribosome stalling mRNA translation [114, 115]. In a prion disease mouse model, GADD34 overexpression alleviates neuronal loss and translation and synaptic impairments [116]. Taken together, emerging research has established a role for ISR in CNS development and homeostasis. Recent findings have also uncovered an important role for ISR in CNS pathologies including neurodegenerative diseases [114, 115].
The role of the ISR in neurodegenerative disorders
Dysregulation of ISR signaling, particularly eIF2α phosphorylation, has been linked to many neurological disorders that are characterized by neuroinflammation, disturbances in protein homeostasis and oxidative stress including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), Huntington disease, vanishing white matter disorder, frontotemporal dementia, neurotrauma, and prion disorders [19, 45]. In neurodegenerative conditions, phosphorylation of eIF2α is a common feature in the canonical adaptive signaling cascade of ISR. Chronic activation of eIF2α contributes to the phenotypic characteristics of neurodegeneration such as neuronal cell death and impaired memory [19]. Downstream signaling of phospho-eIF2α-mediated translation inhibition promotes accumulation of proteins, stress granules, liquid droplets, amyloidogenic processing and neuroinflammation that are associated with neurodegenerative disorders [1, 19, 117,118,119]. Here, we will discuss recent findings on the role and therapeutic potential of ISR in MS, AD, PD and ALS (Fig. 3).
The integrated stress response in neurodegenerative disorders. The integrated stress response (ISR) can be initiated upon sensing ER stress. In Multiple Sclerosis (MS), IFNγ is shown to induce ER stress in myelinating oligodendrocytes (OLs) via the phosphorylation of PERK and eIF2α. The main downstream markers identified in lesions of human MS and the MS animal model EAE (experimental autoimmune encephalomyelitis) are CHOP and BiP. EAE studies show that the ISR can be either detrimental for developing oligodendrocytes or cytoprotective for neurons and mature oligodendrocytes. In Alzheimer’s Disease (AD), the formation of amyloid beta (Aβ) plaques and Tau protein aggregates can initiate ISR primarily through PERK and PKR. ISR markers such as p-PERK and p-eIF2α have been found mainly in the frontal cortex and hippocampus. Evidence implicates PERK activation as either neuroprotective by mitigating Aβ- and tau-induced neuronal death (neuron in green) or cytotoxic by promoting tau phosphorylation and aggregation that lead to neuronal damage (neuron in red). In Parkinson’s Disease (PD), α synuclein and Lewy bodies promote the initiation of the ISR via PERK and HRI phosphorylation. ISR markers are commonly found in the affected substantia nigra and dopaminergic neurons. The PERK-ATF4 signaling pathway is relevant in PD. The involvement of ATF4 in the transcription of Parkin (a pro-survival protein) indicates a cytoprotective role (neuron in green). However, excessive ATF4 activity may contribute to neuronal death by promoting the transcription of pro-apoptotic factors such as Trib3 and CHOP (neuron in red). In Amyotrophic Lateral Sclerosis (ALS) motor neurons in the spinal cord are primarily affected. The ISR is activated by protein aggregation, induced mainly by mutations in TDP-43, SOD1, and FUS. The main markers observed in ALS models and human ALS samples are p-eIF2α, ATF4, CHOP, and BiP. The ISR is mostly detrimental in ALS, and the activation of CHOP in ALS models is linked to apoptosis and neurodegeneration (neuron in red). Figure was created in BioRender
Multiple sclerosis (MS)
MS is an immune-mediated demyelinating disorder of the CNS that results in progressive neurodegeneration [120]. The pathophysiology of MS is characterized by the peripheral activation and infiltration of leucocytes into the CNS, which triggers an inflammatory response and induces the secretion of proinflammatory cytokines that leads to demyelination, oligodendrocyte death, and neurodegeneration [121]. Early reports showed that the ISR-related markers BiP (the expression of which occurs in a constitutive manner), and CHOP (a marker of ER stress) are expressed in post-mortem MS lesions, with a significant increase of CHOP expression in the edges of chronic active lesions [122]. Follow-up studies in the MS animal model, experimental autoimmune encephalomyelitis (EAE) confirmed an increase in the expression of p-eIF2α in EAE lesions [123]. These studies also showed significantly higher expression of CHOP and p-eIF2α in spinal cord neurons of EAE mice.
IFNγ is a key proinflammatory cytokine that drives the pathogenesis of MS and EAE and contributes to demyelination. IFNγ is shown to induce ER stress and promotes apoptosis in developmental rat oligodendrocytes in vitro [124]. Additionally, embryonic mice that received IFNγ ectopically showed hypomyelination, increased expression of the ER-related markers p-eIF2α, BiP, and CHOP, and the apoptosis-related marker Caspase 12 at postnatal day 14 [124]. This evidence indicates a link between hypomyelination, ER stress and IFNγ-induced apoptosis in developmental oligodendrocytes. Further studies in the CNS of adult mice also showed IFNγ induces a moderate ER stress, which activates the ISR via PERK–p-eIF2α ISR in mature oligodendrocytes [125]. However, in this case, the IFNγ-mediated ISR response did not decrease oligodendrocyte survival during EAE. In fact, the activation of the ISR turned out to be protective against EAE-induced demyelination and axonal damage [125]. These two studies show the striking dichotomy in the role of IFNγ-mediated activation of the ISR. While the activation of this pathway is detrimental for developmental oligodendrocytes, it protects mature oligodendrocytes during EAE. This contrasting effect suggests that the IFNγ-mediated ER stress response and the ISR in oligodendrocytes is context-dependent, being detrimental during development but potentially neuroprotective in the context of chronic inflammatory diseases, such as MS.
Interestingly, GADD34-meidtaed dephosphorylation of p-eIF2α is selectively upregulated in myelinating oligodendrocytes of mice that ectopically express IFNγ in the CNS. GADD34-deficient mice at postnatal day 21 also show higher expression of p-eIF2α in oligodendrocytes in the presence of IFNγ, which is associated with reduced oligodendrocyte loss and hypomyelination [126]. These findings demonstrate that enhancing the ISR could promote oligodendrocyte survival in the presence of an MS-relevant cytokine [126]. On the contrary, genetic inactivation of PERK in oligodendrocytes worsens the EAE disease course as 6-week-old PERK-deficient mice show more severe clinical scores and earlier EAE onset with exacerbated demyelination and axonal degeneration compared to their controls [127]. Considering the benefit of ISR activity in oligodendrocytes, further investigation has been conducted to identify the effects of prolonged ISR. Interestingly, these studies show that sustained ISR by prolonging eIF2α phosphorylation can delay the onset of clinical symptoms in 7-week-old EAE mice, reduce oligodendrocyte and axonal loss, and decrease the numbers of T cells in the CNS [128]. More recent findings show that prolonging the ISR can also enhance CNS remyelination in inflammatory conditions. In EAE and cuprizone mouse models of MS, prolonging the ISR protects oligodendrocytes and enhances remyelination. The use of a selective estrogen modulator, bazedoxifene (BZA), increases the number of oligodendrocytes in cuprizone mice, and its combination with Sephin1 (an agent that prolongs ISR) enhanced remyelination [129].
ATF4 is an important downstream effector of the ISR; hence, its role has been investigated in the context of EAE development. Unexpectedly, evidence suggests that specific deletion of ATF4 in oligodendrocytes does not exert discernible impact on EAE severity when compared to their controls [130]. Additionally, ATF4 deficiency in oligodendrocytes does not significantly alter their number, nor changes axon degeneration in EAE lesions. These findings suggest while the PERK-eIF2α pathway protects oligodendrocytes in EAE mice, its downstream effector, ATF4, does not have a significant role in regulating oligodendrocytes’ survival and axonal degeneration during EAE [130]. Recently, stress granules have been shown to exert neuroprotective effects in MS by sequestering pro-apoptotic factors, hence protecting mRNA and proteins from degradation. This was shown in the corpus callosum of cuprizone-induced demyelinating mouse model in which Sephin1 treatment allowed differentiation of oligodendrocyte progenitor cells (OPCs) into oligodendrocytes in the context of inflammatory stress [131]. This study showed the presence of astrocytic IFNγ in GFAP-tTA; TRE-IFN-γ transgenic mice underpins Sephin 1’s effects [131]. Interestingly, Sephin1 extended the IFNγ-triggered ISR by enhancing the levels of p-eIF2α that resulted in reduced protein synthesis and the formation of RNA stress granules in oligodendrocytes [131]. Studies in a 9-week-old EAE mouse model, which recapitulates the inflammatory aspects of MS, have shown suppressing the ISR with 2BAct results in a partial loss of the protective effects of Sephin1 suggesting that Sephin1 may protect the CNS from inflammation by enhancing the ISR. These findings have provided initial evidence that Sephin1 may protect oligodendrocytes through the formation of RNA stress granules [131]. Although these EAE findings aid in understanding the role of the ISR in MS, these studies have their limitations. MS is a disease that progresses over time, and its severity can increase with age. Thus, EAE findings fail to address the relevance of the ISR in progressive MS. Further investigation in chronic progressive models of MS would allow uncovering the role of ISR in disease progression and aging in MS. Future research will also need to focus on understanding how ISR modulate activity of mature and older oligodendrocytes, and its role in remyelination.
Recent studies from active and inactive human MS lesions have also shown the presence of stress granules in oligodendrocytes, which has been also confirmed in cultures of human oligodendrocytes under metabolic stress [132]. These studies revealed that stress granules persist in oligodendrocytes while pro-inflammatory cytokines are present in culture media [132]. Interestingly, while PERK activity is shown to be important for oligodendrocytes under injury, neither PERK activation nor deletion alters the viability or myelinating capacity of oligodendrocytes under homeostasis [127, 133]. Similarly, the conditional deletion of ATF4 in oligodendrocytes has shown negligible effects in the CNS under homeostasis; with little effect on myelination, and a similar number of oligodendrocytes and axons between the ATF4 conditional knockout and the wild-type mice [130]. Altogether, emerging evidence suggests the involvement of ISR pathways in regulating oligodendrocytes in MS; however, further research is required to elucidate the underlying mechanisms by which ISR contributes to MS pathogenesis and progression. Future research is required to elucidate the underlying mechanisms by which mitochondrial dysfunction (a known mechanism of MS pathogenesis) [134,135,136] engages in a crosstalk with ISR during MS progression and its pathogenesis.
The role of ISR in Alzheimer’s Disease (AD)
AD is a neurodegenerative condition commonly characterized by memory loss, depression, disorientation, behavioral changes, and motor-related impairments [137]. AD causes synaptic and neuronal loss, oxidative damage, neuroinflammation, among others. Plaques of amyloid-beta (Aβ) and neurofibrillary tangles (NFTs), which are phosphorylated aggregates of tau protein, are hallmarks of AD. As reviewed previously, Aβ can disturb the function of proteasomes and lysosomes, impair calcium homeostasis, and enhance the formation of NFTs in neurons [138]. Furthermore, the continuous accumulation of Aβ and phosphorylated tau can result in abnormal protein folding and subsequent ER stress [138]. Early studies on post-mortem AD brain showed upregulation of p-PERK and p-eIF2α in the hippocampus and frontal cortex. While the results of this study were initially associated with the UPR only, it is important to consider that the ISR and the UPR converge in the phosphorylation of PERK and eIF2α, which may suggest the involvement of both pathways [139]. Subsequent studies established a relationship between tau phosphorylation and ER stress. Higher expression of p-PERK and p-eIF2α were reported in the hippocampus of P301L aged mice, a transgenic mouse model that develops tau pathologies [140]. Additionally, primary cortical neurons from embryonic day 17 Sprague–Dawley rats, treated with the ER-stressor thapsigargin showed tau hyperphosphorylation and cleavage. Interestingly, inducing tau hyperphosphorylation in vitro with okadaic acid led to increased immunoreactivity of p-PERK accompanied by a significant increase in p-eIF2α [140]. Consistent with these findings, studies in 6-month-old P301L mice showed increased expression of p-eIF2α and ATF4, and low protein synthesis compared to wild-type mice [141]. Interestingly, the levels of spliced XBP1 and ATF6 did not increase at the 6-month mark, which rules out the involvement of the UPR and suggests the presence of ISR signaling. Additionally, it was found that activation of PERK-eIF2α contributed to the pathological phosphorylation of tau in rTg4510 mice, a mouse model of tau pathology with pronounced neurodegeneration similar to human tauopathies. Contribution of PERK-eIF2α axis to tau pathology was further supported when suppressing PERK activity in P301L mice with the PERK-specific inhibitor GSK2606414 restored the rate of protein synthesis by downregulating p-PERK, p-eIF2α, and ATF4. Furthermore, P301L mice treated with GSK2606414 show a significantly higher number of CA1 pyramidal neurons and ameliorate brain atrophy suggesting PERK inhibition may provide neuroprotection [141].
Other studies suggest that PERK-eIF2α signaling might be beneficial in AD [142]. Knockdown of PERK in SK-N-SH human neuroblastoma cell lines enhances Aβ-mediated cell death in neurons by suppressing eIF2α and the Grp78/Bip ER chaperon [142]. Promoting PERK signaling is also shown to mitigate tau pathologies both in vivo and in vitro [143]. In an in vitro model of tauopathy in human neurons, enhancing PERK signaling through pharmacological and genetic approaches reduces tau phosphorylation and tau conformational changes that promote neuronal survival. Moreover, PERK activation significantly improves memory and locomotion in P301S tau mice by reducing tau pathology, promoting dendritic spine density and attenuating motoneuron loss [143]. Human studies have identified the presence of EIF2AK3 variants (encoding for PERK) in Dutch patients with AD, indicating that these variants may result in an increased risk of developing the disease [144]. Follow-up studies have further demonstrated that PERK has a role in tau protein aggregation. In vitro assessments indicate that inhibiting PERK directly (hence decreasing eIF2α phosphorylation) promotes tau aggregation and increasing eIF2α phosphorylation prevents tau aggregation in Biosensor cells. In accordance with these findings, post-mortem brain samples from AD donors show that PERK signaling is downregulated in the hippocampus [145].
Other EIF2A kinases have been implicated in AD. Early studies showed that Aβ peptides promote PKR phosphorylation in rat primary neurons and suggested that increased intracellular calcium is important for the Aβ peptide-activation of the PKR-eIF2α pathway [146]. Recently it has been proposed that PKR may directly regulate tau expression in AD as in vitro assessments on OLN-T40AS cells showed that PKR directly phosphorylates several abnormal residues within tau [147]. Furthermore, changes in PKR expression led to corresponding changes tau mRNA and protein levels in OLN-T40AS cells. Brain slices from the AD rTg4510 mouse model treated with a PKR inhibitor show reduced phosphorylation of soluble tau [147]. GCN2 has also been investigated; however, evidence shows that this kinase does not have a significant role in AD. Findings from 5XFAD mice (a mouse model of severe amyloid pathology) with GCN2 deletion suggest that overactivation of PERK facilitates phosphorylation of eIF2α, and aggravates the expression of hippocampal BACE1 and ATF4, failing to rescue memory deficits in a fear conditioning test [116].
In AD, an imbalance in mitochondrial proteastasis due to accumulation of unfolded proteins in the mitochondrial matrix is shown to increase the expression of key genes that are involved in stabilization of mitochondria during Aβ-mediated proteotoxicity [148]. In one such study, Beck and colleagues showed that UPRmt is prominently activated in frontal cortex of both sporadic and familial AD human postmortem tissue samples [149]. Another study by Shen and colleagues in APPsw/PS1dE9 transgenic mice and SH-SY5Y cell line revealed that an increase in the expression of UPRmt proteins such as Hsp60, CLPP and Htr/Omi is observed in 3-month-old mice while remained unchanged in 9-month-old mice comparing their age-matched wildtype-control [65]. This study suggests that activation of UPRmt decreases with aging during the process of mitochondrial dysfunction in AD [65]. The authors also found that sphingolipid biosynthesis and mevalonate pathways are necessary for the activation of UPRmt induced by Aβ, and inhibiting these pathways in SH-SY5Y cells prevents UPRmt activation, aggravates abnormal mitochondrial structure, increases ROS levels, and exacerbates cytotoxicity mediated by Aβ plaques. These changes are known to have modulatory effects on ISR, limiting the beneficial effects of UPRmt. Taken together, growing evidence from preclinical studies have demonstrated a link between the ISR and AD pathology.
Preclinical AD models have been crucial to elucidate the role of ISR in AD and to assess how its inhibition with ISRIB would impact AD pathophysiology. Current evidence from mouse models suggests a controversial role for ISR in AD. For instance, there are animal studies that show contrasting effects of ATF-4 in mediating Aβ plaque induced neuropathology. An intra-axonal translation of ATF-4 was reported to induce Aβ1-42 mediated neurodegeneration in a retrograde manner [113, 150]. Another work also showed that ISRIB treatment prevents Aβ-induced cell death in the neuronal cell line PC12 [151]. These studies proposed that this effect is likely through the inhibition of ATF4 with no impact on Aβ production [151]. In contrast, recent studies have identified that the small molecule ISRIB provides neuroprotection against the disruptive effects of Aβ on synaptic integrity and cognition in a rat model of sporadic AD [152]. It was suggested that this effect may be due to a restoration of Aβ-induced aberrant protein synthesis and increased expression of ATF4 in the hippocampus [152]. There are other reports that also demonstrate the beneficial effects of PERK and eIF2α phosphorylation in tau pathology [142, 143]. Overall, these examples suggest a controversial role of ISR in AD pathology that requires further investigation.
To critically interpret these conflicting reports, it is crucial to consider that these studies have their limitations. Firstly, current AD models do not fully replicate the complex pathophysiology of this disease. As an age-related neurodegenerative disease, it is important to consider that most animal models have short lifespans, which limits the ability to effectively mimic disease progression. Furthermore, findings related to the protective effects of ISR activation observed in these models could be a secondary effect to the artificial overexpression of proteins. Understanding the dynamic interplay between disease progression and its relationship with different ISR kinases, and cellular stress responses are among the critical points to address. Additionally, the timing and duration of the ISR can impact tau pathology and AD progression. Overall, due to the complexity of AD, further investigation on the effects of pharmacological modulation on the ISR and its different branches at different stages would help elucidate the mechanisms of this pathology and help identify potential therapeutic approaches.
The ISR and Parkinson’s Disease (PD)
PD is another neurodegenerative disease that impacts dopaminergic neurons in the substantia nigra causing disruption in nigrostriatal dopaminergic innervation in the brain. PD is characterized by the misfolding aggregation of α-synuclein that contributes to the formation of Lewy bodies, a hallmark of this disease [153].The formation of these aggregates, along with an impaired protein clearance by the ubiquitin–proteasome and autophagy-lysosomal systems, mitochondrial dysfunction, neuroinflammation, and oxidative stress collectively contribute to neurodegeneration [154]. Another key aspect of this disease is the failure of mitochondrial quality control, which plays a major role in neuronal survival. Mitophagy, the process where excessive or damaged mitochondria are degraded by lysosomal hydrolases, is crucial for maintaining homeostasis [155]. Recent findings demonstrated that the HRI branch of the ISR selectively induces mitophagy via the mitochondrial localization of peIF2α [156]. In this study, K562 engineered cells, which express the mitophagy reporter mito-mKeima, were treated with either the BTdCPU or Salubrinal. Flow cytometry identified an increase in HRI-dependent mitophagy, and both treatments were associated with the accumulation of ATF4 and p-eIF2α protein levels [156]. Additionally, it was reported that the HRI mitophagy pathway is activated in parallel with the mitophagy pathway regulated by the PD-associated genes PINK1 and PARKIN, with distinct underlying mechanisms. Therefore, it is suggested that the HRI pathway, which normally results in translational initiation, can trigger mitophagy in response to mitochondrial damage [156]. Although these are promising findings that could be associated with a mechanism of PD, it should be noted that the study was limited to K562 and HeLa cell lines, which are not relevant in vitro PD models. Further studies with PD cell lines will be required to confirm an association between HRI-dependent mitophagy and neuronal integrity.
PD is part of a group of diseases known for abnormal proteostasis and accumulation of misfolded proteins that result in neuronal dysfunction [157]. ER stress and possibly ISR have been associated with PD. Early studies on post-mortem brain samples identified the presence of p-PERK and p-eIF2α in the substantia nigra of PD brain in which 5–20% of the α-synuclein positive neurons were immunoreactive for p-PERK while it was absent in the control non-PD brain [158]. Moreover, over-expression of truncated α-synuclein in HEK 293 cell line, which does not express wild-type α-synuclein, results in high expression of GRP78/BiP. Interestingly, under glucose deprivation, dopaminergic neurons differentiated from SH-SY5Y cells have shown formation of intracellular aggregates of α-synuclein, and some of these aggregates expressed GRP78/BiP and ATF4 [159]. In agreement with these findings, dopaminergic neurons of the substantia nigra in SYN120 transgenic mice (a model of PD that overexpresses α-synuclein) express higher levels of GRP78/BiP as compared to their wildtype controls [15, 159]. Altogether, in vitro and in vivo evidence from PD models suggest the involvement of PERK signaling of the ISR in the disease.
Studies by Bouman and colleagues reported that ATF4 is involved in the transcription of Parkin, a pro-survival protein that acts as a regulator for protein breakdown and mitochondrial integrity [160]. In this study, SH-SY5Y cells and primary mouse cortical neurons with mitochondrial damage showed increased transcription of Parkin and BiP mRNA. Furthermore, Parkin is upregulated in response to thapsigargin- and tunicamycin-induced ER stress and is identified as a downstream target of the PERK/ATF4 pathway [160]. Interestingly, ATF4 knockdown in vitro can significantly reduce expression of Parkin. This study determined that Parkin expression upon PERK activation seems to be neuroprotective due to its role in reducing mitochondrial damage [160]. In support of this evidence, another study has identified that ATF4 has a cytoprotective role in PD [161]. Silencing ATF4 promotes cell death of neuronal PC12 cells in the presence of dopaminergic neurotoxins 6-OHDA and MPP + [161]. Conversely, ATF4 overexpression reduces cell death by maintaining the expression of Parkin [161]. Although these studies showed a protective role of PERK/ATF4, a follow-up study from the same group reported that CHOP and ATF4 mediate the transcription of Trib3 (tribbles pseudo kinase 3), a protein with pro-apoptotic function that is highly activated in PC12 cells under 6-OHDA in an in vitro model of PD [162]. These in vitro results were confirmed in human post-mortem PD brain where Trib3 was highly expressed in nigral dopaminergic neurons [162]. Furthermore, sustained over-expression of ATF4 by rAAV in a rat model of PD-like neurodegeneration, induced by human wild type α-synuclein, is shown to promote neuronal cell death in the substantia nigra pars compacta (SNpc) [163]. Studies in a mouse of model PD-like progression reported that ATF4 binds to the E3 ligase parkin promoter thereby regulating its expression in response to mitochondrial and ER stress [164]. These findings show that the increased expression of parkin due to ATF4 is beneficial during mitochondrial damage and ER stress as PD is developed. The brain biopsy tissues from PD patients also exhibit elevated levels of ATF-4 immunostaining in the neuromelanin-positive neurons suggesting that ATF-4 expression is affected by the mean duration of the PD; the longer the progression the higher the ATF-4 expression levels [160].
The synucleinopathy is also known to disrupt the mitochondrial health through dysfunctional membrane potential, degeneration of mitochondrial complex I, disruption of Ca2+ homeostasis, and an increased release of cytochrome c [165]. Moreover, an imbalance in mitochondrial proteostasis due to soluble oligomers of prefibrillar α-synuclein is commonly observed in PD [166]. Other known factors that disturb mitochondrial homeostasis in PD include disruption of retromer complex which results in mitochondrial fragmentation due to mutation in VPS35 gene [167]; deficiency of mitochondrial protein kinase; and PTEN-induced putative (PINK)1 which is associated with an autosomal recessive variant (PARK6) of PD [168].
Therapeutically, in efforts to target PERK-ATF4 axis in PD, oral administration of the small molecule GSK2606414 to mice models of PD has resulted in the inhibition of PERK activity in the SNpc after experimental ER stress stimulation with tunicamycin [157]. The inhibitor attenuates 6-OHDA mediated cell death of nigral-dopaminergic neurons in PD mice, which improves motor performance and recovery of dopamine and two synaptic proteins, VAMP (Vesicle-associated membrane protein 2) and SNAP25 (Synaptosomal-Associated Protein, 25 kDa) [157]. A recent study in mouse mesencephalic and cortical neuronal cultures has demonstrated that treatment with PD-relevant neurotoxins, MPP + and 6-OHDA, as well as α-synuclein aggregation results in the sustained expression of ATF4 in the nucleus of dopaminergic neurons [169]. These neurotoxins also promote the expression of proapoptotic factors CHOP, Trib3, and Puma through ATF4 activity in dopaminergic neurons. Interestingly, pharmacological inhibition of PKR in cortical and dopaminergic neurons with imidazole-oxindole or C16 suppressed ATF4 [169]. Furthermore, ATF4 inhibition did not reduce the PD neurotoxin-induced eIF2α phosphorylation that suggests ATF4 regulation is independent of PKR, signifying the role of the PERK-ATF4 signaling in PD [169].
In summary, emerging evidence has identified the ISR as a potential therapeutic target for PD. However, further research is warranted to decipher ISR mechanisms in PD, particularly the controversial findings on the involvement of PERK-ATF4 pathway in both neurodegeneration and neuroprotection processes in PD. Like AD, preclinical models of PD rely on genetic and/or pharmacological manipulation to mimic this pathophysiology in rodents. Hence, it may be argued that the effects of the ISR on neurons could be an artificial response to these stimuli. Given that these current experimental models may not fully replicate the mechanisms of ISR in PD pathology, clinical studies are necessary to provide more insight into the positive or negative effects of the ISR.
The ISR in Amyotrophic Lateral Sclerosis (ALS)
ALS is a devastating neurodegenerative disease that results in the loss of motor neurons in the CNS and is associated with axonal retraction and neuromuscular denervation. ALS has pathological heterogeneity as it can be caused by mutations in SOD1 (Cu–Zn superoxide dismutase) and FUS (fused in sarcoma) genes, which result in the formation of protein aggregates with different compositions [170]. The common signature of ALS is the aggregation of TDP-43 (TAR DNA-binding protein 43) [171]. TDP-43 is a nuclear RNA/DNA binding protein encoded by the TARDBP gene with important roles in transcriptional regulation, alternative splicing, and mRNA stabilization [171, 172]. With motor neurons injury, TDP-43 is released from the nucleus and its mis-localization aids in forming intracellular aggregates in the cytoplasm that is a hallmark of ALS pathology [171]. Compared to other neurodegenerative disorders, the ISR has been well described in ALS. Early studies on human ALS samples identified upregulation of ISR markers, such as p-eIF2α, in spinal cord neurons [173]. Development of different animal models of ALS has also aided in understanding the role of ISR signaling in regulating motor neurons in ALS. The mutant SOD1 mouse model that mimics familial ALS is known to cause protein misfolding and aggregates formation, thus it is commonly used to study the role and mechanisms of the ISR in ALS complementing human ALS studies. Evidence also suggests high expression of CHOP in neurons, oligodendrocytes, microglia, and astrocytes in the Rexed laminae IX anterior horn of the spinal cord of sporadic ALS patients and the SOD1G93A mouse [173]. Overall, emerging animal studies have provided supportive evidence that ER stress, UPR and the ISR have an important role in ALS pathophysiology.
Given the diversity of genetic mutations that can cause ALS, it is proposed that the ISR can be triggered through different mechanisms. Studies in SOD1 mutants have suggested accumulation of different SOD1 mutant proteins can induce UPR by phosphorylation of IRE1 and PERK [174]. Furthermore, the interaction between the protein Derlin-1 and SOD1 mutants has been proposed as an underlying mechanism for inducing ER stress and UPR activation [174]. Higher levels of ATF6 and p-eIF2α in the motor neurons of transgenic SOD1G93A mice has further substantiated the involvement of UPR in ALS, although the levels of spliced XBP1 or sXBP1 (a downstream marker of the UPR/IRE1 pathway) are not significantly different [175]. These studies proposed that ATF6 activation may occur earlier in ALS than XBP1 by proteolysis, and the latter is upregulated upon de novo synthesis [175]. Interestingly, a high expression level of UPR-related markers like sXBP1, p-eIF2α, GRP78/BiP, and CHOP are also detected in the skeletal muscle of SOD1G93A mice, suggesting that the UPR pathway may be implicated in the muscle atrophy and weakness observed in ALS by affecting muscle cells instead of direct effects on motor neurons and motor pathways [176].
Studies in SOD1 mouse model have reported different activation profile of UPR system based on the type of transgenic mice. Although motor neurons of transgenic SOD1G93A mice have shown increased UPR activity via PERK and ATF6, transgenic SOD1G85R mouse model only showed significant activation of PERK signaling in motor neurons through upregulation of ATF4 and CHOP with no changes in the levels of BiP and sXBP1 [177]. Given the heterogeneity of ALS, it is plausible that some mutations result in UPR activity while others activate the ISR, without involving the UPR. A genome-wide screening for activators and suppressors of the ALS-associated gene C9ORF72A using the CRISPR Cas9 system supports this hypothesis by identifying a robust activity of the ISR, but not the UPR [178]. More recent findings demonstrated that patients with C9ORF72 repeat expansion mutations show increased levels of phosphorylated PKR, suggesting that other ISR-related eIF2α kinases are involved in ALS [179]. Further studies from a Drosophila model and in rat primary neurons have also provided evidence that the PERK inhibitor GSK2606414 reduces TDP43 toxicity, supporting a role for the ISR in neurotoxicity [180]. SOD1G85R/GADD34+/ΔC mice with a mutation in GADD34 show an enhanced phosphorylation of eIF2α in spinal cord homogenates, indicating prolonged ISR [181]. Additionally, SOD1G85R/GADD34+/ΔC mice show a delayed onset of the disease with higher survival rates when compared to the SOD1G85R control. Thus, it has been proposed that the beneficial effects observed in the SOD1G85R/GADD34+/ΔC are attributed to an enhanced ISR that delays accumulation and formation of mutant SOD1 aggregates.
Evidence from three different familial ALS mouse models has helped classify two types of motor neurons according to their vulnerability or resistance to ER stress [182]. Interestingly, fast-fatigable motor neurons are more vulnerable and prone to developing ER stress and show an increased expression of p-eIF2α, while the fatigue-resistant neurons demonstrate a delayed response to ER stress [182]. This work provided a mechanism underpinning early disease manifestations related to the susceptibility of motor neurons, and late manifestations in the resistant neurons [182], and it might indicate a relationship between the ISR and highly active motor neurons with high metabolic demands. Recent work has provided more insight into the role of the ISR by using the rNLS8 mouse model, which mimics aggregates of insoluble cytoplasmic TDP-43 observed in ALS [183]. Transcriptomic and protein analyses of rNLS8 mice indicate cortical upregulation of ISR- and apoptosis-related genes ATF4, CHOP, Bid, Gadd45, Trp53 before the disease onset. Upregulation of CHOP at the onset of the disease and increased levels of cleaved caspase-3 in the cortical neurons of rNLS8 mice suggested a role for CHOP in apoptosis-mediated neurodegeneration in ALS. However, further evidence showed that the knockdown of CHOP in rNLS8 mice did not alter the TDP-43 pathology and caspase-3 activation, suggesting other pathways that may account for activation [183]. Intriguingly, a recent study has suggested that the ISR might have a protective rather than pro-apoptotic role in ALS [184]. This in vitro study on human P525L FUS neurons indicate that FUS mutations result in the activation of the heat-shock response and the ISR [184]. However, cell survival was similar in mutant and wild-type neurons under various stress conditions. This suggests that over-activation of stress response pathways in mutant cells may protect them from susceptibility to cell death. However, when the ISR was acutely inhibited under stress conditions the outcome did not change, suggesting that the ISR on its own may not be responsible for these protective effects. Altogether, while further studies are required to clarify these findings, it is plausible that the ISR may act as an early protective mechanism in response to the accumulation of FUS in the cytoplasm by preventing cell death, however, it can turn into a toxic mechanism over time [184].
The ER-mitochondrial crosstalk is also known to regulate synaptic transmission, and its impairment can lead to dysfunctional synapses which is observed in many neurodegenerative disorders like ALS [185]. Accumulating evidence also indicates that mitochondrial dysfunction plays an important role in ALS pathogenesis, as ER-mitochondrial physical and functional connection contributes to the calcium homeostasis and lipids biosynthesis [186]. These reports have indicated a pivotal role for SOD1 mediated mitochondrial dysfunction. Recent evidence in familial ALS cases has also unraveled that mutations like TARDBP and C9ORF72 contribute to morphological defects in mitochondria [187]. The role of mitochondria and ER crosstalk in modulation of ISR during ALS pathogenesis requires further investigations.
Preclinical research has been conducted to develop therapeutic strategies for modulating the ISR in ALS. Guanabenz, a non-selective inhibitor of ER stress, has been tested in ALS. In animal models of ALS, Guanabenz, that is commonly used to treat hypertension, has been identified to induce eIF2α dephosphorylation and persistent activity of the ISR, allowing the clearance of misfolded proteins and prolonging motor neurons survival [188]. A multicentre, randomized, double-blind phase 2 clinical trial has reported that guanabenz at two doses, 64 and 32 mg, can slow the progression of ALS in patients with bulbar onset, which is the most homogenous phenotype of ALS [189]. DNL343 is another candidate treatment that has been investigated in A recent phase I trial in patients with ALS for its potential as eIF2B agonist. DNL343 is shown to attenuate the formation of ISR stress granules in vitro, [190]. The results of the phase I trial have identified the safety of DNL343 in humans; however, its efficacy is yet to be tested in future phase II trials.
Conclusions and future perspectives
The integrated stress response has gained increasing attention in the past two decades for its role in pathogenesis and progression of neurodegenerative diseases. There is now more understanding of how the eIF2α kinases detect various types of stress and converge on the phosphorylation of eIF2α to regulate the CNS [116]. It is also increasingly acknowledged that the ISR is a critical pathway that ensures cells can respond to stress, maintain proteostasis, and adapt to changing conditions. The ISR also plays pivotal role in maintaining CNS homeostasis, particularly during neurodevelopment and memory consolidation, where various eIF2α kinases and downstream signaling pathways collectively contribute to the intricate regulation of synaptic plasticity in the brain [45].
Given the important role of ISR in regulating proteostasis, learning, and memory formation, there has been growing interest in identifying mechanisms that link the ISR and neurodegenerative disorders, where proteotoxicity and cognition decline are involved. Recent animal and human studies have confirmed the involvement of ISR in pathogenesis of various neurodegenerative disorders. Hence, efforts have been made to develop therapeutic strategies to target ISR pathways. In MS preclinical studies, where demyelination and oligodendrocyte loss are the hallmarks of the disease, current evidence suggests that prolonging the ISR with Sephin1 might be a potential strategy to promote remyelination [128, 129, 131]. In AD, pharmacologic and genetic enhancement of PERK signaling has shown promise to reduce tau phosphorylation [143]. Interestingly, PKR enhancement results in tau phosphorylation, while its inhibition has the opposite effect [147]. These findings suggest the complexity of ISR signaling and the differential response of various kinases in the pathway, signifying the importance of this information for developing therapeutic approaches. In PD, the PERK-ATF4 pathway has been identified as a potential target since inhibiting PERK activity with GSK2606414 has shown neuroprotective effects and preserves dopaminergic neurons in PD animal models [157]. The ISR has been studied more extensively in ALS and Guanabenz is currently being evaluated as a therapeutic strategy that has shown encouraging results in a phase II clinical trial [189].
In conclusion, ISR has emerged as a promising target in neurodegenerative diseases. Our current understanding of cellular communication of stress responses to mount a cytoprotective role (at single cell level) or to mitigate the overall stress in a collective cellular context is a subject of further investigation. Thus, it is essential to develop insights into the outcomes of targeting the stress response pathways by modulating the ISR in different cell types and disease phenotypes. This knowledge is key to uncovering the regulatory mechanisms of different stress responses across the organism and would facilitate development of effective pharmacological based treatments. To this end, the timing of ISR activation during the development of neurodegenerative phenotypes and linking these events with other cellular pathways such as mitophagy, oxidative stress, ER stress, immunomodulatory response, and metabolism are among the most crucial pathways to unravel. Another major knowledge gap in this area is how the pro-survival and pro-apoptotic pathways are mediated by the eIF2α kinases or the underlying mechanisms that direct the preferential translation of certain mRNAs. Altogether, further understanding of the dual roles that ISR play in survival and apoptosis is crucial for developing effective therapeutic interventions to modulate the ISR in these neurodegenerative conditions.
References
Pakos-Zebrucka K, et al. The integrated stress response. EMBO Rep. 2016;17:1374–95.
Lin JH, Walter P, Yen TSB. Endoplasmic reticulum stress in disease pathogenesis. Ann Rev Pathol: Mech Dis. 2008;3:399–425. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.pathmechdis.3.121806.151434.
Balachandran S, et al. Essential role for the DsRNA-dependent protein kinase PKR in innate immunity to viral infection. Immunity. 2000;13:129–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s1074-7613(00)00014-5.
Han AP, et al. Heme-regulated eIF2alpha kinase (HRI) is required for translational regulation and survival of erythroid precursors in iron deficiency. EMBO J. 2001;30:6909–18.
Deegan S, et al. A close connection between the PERK and IRE arms of the UPR and the transcriptional regulation of autophagy. Biochem Biophys Res Commun. 2015;456:305–11.
Kapuy O. Mechanism of decision making between autophagy and apoptosis induction upon endoplasmic reticulum stress. Int J Mol Sci. 2024;25. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms25084368.
Rouschop KMA, et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J Clin Investig. 2010;120:127–41.
Liu CY, Schroder M, Kaufman RJ. Ligand-independent dimerization activates the stress response kinases IRE1 and PERK in the lumen of the endoplasmic reticulum. J Biol Chem. 2000;275:24881–5.
Kazemi S, et al. A novel function of eIF2α kinases as inducers of the phosphoinositide-3 kinase signaling pathway. Mol Biol Cell. 2007;18:3635–44.
Hu J. et al. Activation of ATF4 mediates unwanted Mcl-1 accumulation by proteasome inhibition. 2012. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood-2011.
Zhang G, et al. Integrated stress response couples mitochondrial protein translation with oxidative stress control. Circulation. 2021;144:1500–15.
Clementi E, et al. Persistent DNA damage triggers activation of the integrated stress response to promote cell survival under nutrient restriction. BMC Biol. 2020;18-36.
Alagar Boopathy LR, Jacob-Tomas S, Alecki C, Vera M. Mechanisms tailoring the expression of heat shock proteins to proteostasis challenges. J Biol Chem. 2022;298. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jbc.2022.101796.
Fu Y, et al. Mitochondrial DNA breaks activate an integrated stress response to reestablish homeostasis. Mol Cell. 2023;83:3740-3753.e9.
Donnelly N, Gorman AM, Gupta S, Samali A. The eIF2α kinases: Their structures and functions. Cell Mol Life Sci. 2013;70:3493–511. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-012-1252-6.
Sidrauski C, McGeachy AM, Ingolia NT, Walter P. The small molecule ISRIB reverses the effects of eIF2α phosphorylation on translation and stress granule assembly. Elife. 2015;4:e05033.
Han J, et al. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat Cell Biol. 2013;15:481–90.
Wortel IMN, van der Meer LT, Kilberg MS, van Leeuwen FN. Surviving stress: modulation of ATF4-mediated stress responses in normal and malignant cells. Trends Endocrinol Metab. 2017;28:794–806. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tem.2017.07.003.
Bond S, Lopez-Lloreda C, Gannon PJ, Akay-Espinoza C, Jordan-Sciutto KL. The integrated stress response and phosphorylated eukaryotic initiation factor 2α in neurodegeneration. J Neuropathol Exper Neurol. 2020;79:123–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jnen/nlz129.
Taniuchi S, Miyake M, Tsugawa K, Oyadomari M, Oyadomari S. Integrated stress response of vertebrates is regulated by four eIF2α kinases. Sci Rep. 2016;6:32886. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep32886.
Colthurst DR, Campbell DG, Proud CG. Structure and regulation of eukaryotic initiation factor eIF-2: Sequence of the site in the α subunit phosphorylated by the haem-controlled repressor and by the double-stranded RNA-activated inhibitor. Eur J Biochem. 1987;166:357–63.
Pathak VK, Schindler D, Hershey’ JWB. Generation of a mutant form of protein synthesis initiation factor EIF-2 lacking the site of phosphorylation by EIF-2 kinases. Mol Cell Biol. 1988;8:993-5.
Liang SH, Zhang W, McGrath BC, Zhang P, Cavener DR. PERK (eIF2α kinase) is required to activate the stress-activated MAPKs and induce the expression of immediate-early genes upon disruption of ER calcium homoeostasis. Biochem J. 2006;393:201–9.
Cullinan SB, Diehl JA. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J Biol Chem. 2004;279:20108–17.
Liu Z, Lv Y, Zhao N, Guan G, Wang J. Protein kinase R-like ER kinase and its role in endoplasmic reticulum stress-decided cell fate. Cell Death Dis. 2015;6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cddis.2015.183.
Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol. 2000;2. www.nature.com/ncb.
Carrara M, Prischi F, Nowak PR, Kopp MC, Ali MM. Noncanonical binding of BiP ATPase domain to Ire1 and Perk is dissociated by unfolded protein CH1 to initiate ER stress signaling. 2015 https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.03522.001
Wang P, Li J, Tao J, Sha B. The luminal domain of the ER stress sensor protein PERK binds misfolded proteins and thereby triggers PERK oligomerization. J Biol Chem. 2018;293:4110–21.
Volmer R, Van Der Ploeg K, Ron D. Membrane lipid saturation activates endoplasmic reticulum unfolded protein response transducers through their transmembrane domains. Proc Natl Acad Sci USA. 2013;110:4628–33.
De La Cadena SG, Hernández-Fonseca K, Camacho-Arroyo I, Massieu L. Glucose deprivation induces reticulum stress by the PERK pathway and caspase-7- and calpain-mediated caspase-12 activation. Apoptosis. 2014;19:414–27.
Nakamura T, et al. Double-stranded RNA-dependent protein kinase links pathogen sensing with stress and metabolic homeostasis. Cell. 2010;140:338–48.
Onuki R, et al. An RNA-dependent protein kinase is involved in tunicamycin-induced apoptosis and Alzheimer’s disease. EMBO J. 2004;23:959–68.
Li G, Scull C, Ozcan L, Tabas I. NADPH oxidase links endoplasmic reticulum stress, oxidative stress, and PKR activation to induce apoptosis. J Cell Biol. 2010;191:1113–25.
Ukhueduan B, Chukwurah E, Patel RC. Regulation of PKR activation and apoptosis during oxidative stress by TRBP phosphorylation. Int J Biochem Cell Biol. 2021;137:106030.
Sharma V, et al. eIF2α controls memory consolidation via excitatory and somatostatin neurons. Nature. 2020;586:412–6.
Wek SA, Zhu S, Wek RC. The Histidyl-TRNA synthetase-related sequence in the EIF-2 protein kinase GCN2 interacts with TRNA and is required for activation in response to starvation for different amino acids. Mol Cell Biol. 1995;15:4497–506.
Mazroui R, Marco SD, Kaufman RJ, Gallouzi I-E. Inhibition of the ubiquitin-proteasome system induces stress granule formation □ D. Mol Biol Cell. 2007;18:2603–18.
Jiang HY, Wek RC. Phosphorylation of the α-subunit of the eukaryotic initiation factor-2 (eIF2α) reduces protein synthesis and enhances apoptosis in response to proteasome inhibition. J Biol Chem. 2005;280:14189–202.
Deng J, et al. Activation of GCN2 in UV-irradiated cells inhibits translation vated protein (MAP) kinase signaling modules to elicit additional transcriptional responses to UV irradiation [6–8]. Curr Biol. 2002;12:1279-86.
Mukherjee T, et al. The eIF2α kinase HRI triggers the autophagic clearance of cytosolic protein aggregates. J Biol Chem. 2020;296:100050.
Abdel-Nour M, et al. The heme-regulated inhibitor is a cytosolic sensor of protein misfolding that controls innate immune signaling. Science. 2019;365(6448):eaaw4144.
Patel S, et al. Cytoplasmic proteotoxicity regulates HRI-dependent phosphorylation of eIF2α via the Hsp70-Bag3 module. iScience. 2022;25(5):104282.
Jackson RJ, Hellen CUT, Pestova TV. The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol. 2010;11:113–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm2838.
Kimball SR. Eukaryotic initiation factor eIF2. Int J Biochem Cell Biol. 1999;31:25–9.
Costa-Mattioli M, Walter P. The integrated stress response: From mechanism to disease. Science (New York, N.Y.) 2020;368. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.aat5314.
Podust LM, Krezel AM, Kim Y. Crystal structure of the CCAAT box/enhancer-binding protein beta activating transcription factor-4 basic leucine zipper heterodimer in the absence of DNA. J Biol Chem. 2001;276:505–13.
Liu G, et al. Salermide up-regulates death receptor 5 expression through the ATF4-ATF3-CHOP axis and leads to apoptosis in human cancer cells. J Cell Mol Med. 2012;16:1618–28.
B’Chir W, et al. The eIF2α/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Res. 2013;41:7683–99.
Han HE, Kim TK, Son HJ, Park WJ, Han PL. Activation of autophagy pathway suppresses the expression of iNOS, IL6 and cell death of LPS-stimulated microglia cells. Biomol Ther (Seoul). 2013;21:21–8.
Halterman MW, De Jesus C, Rempe DA, Schor NF, Federoff HJ. Loss of c/EBP-β activity promotes the adaptive to apoptotic switch in hypoxic cortical neurons. Mol Cell Neurosci. 2008;38:125–37.
Huggins CJ, et al. C/EBPγ is a critical regulator of cellular stress response networks through heterodimerization with ATF4. Mol Cell Biol. 2016;36:693–713.
Hu H, Tian M, Ding C, Yu S. The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front Immunol. 2019;10. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2018.03083.
Novoa I. et al. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of EIF2. J Cell Biol. 2001;153 http://www.jcb.org/cgi/content/full/153/5/1011.
Jousse C, et al. Inhibition of a constitutive translation initiation factor 2α phosphatase, CReP, promotes survival of stressed cells. J Cell Biol. 2003;163:767–75.
Eckl EM, Ziegemann O, Krumwiede L, Fessler E, Jae LT. Sensing, signaling and surviving mitochondrial stress. Cell Mol Life Sci. 2021;78:5925–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-021-03887-7.
Baker BM, Nargund AM, Sun T, Haynes CM. Protective coupling of mitochondrial function and protein synthesis via the eIF2α kinase GCN-2. PLoS Genet. 2012;8:e1002760.
Eckl E-M, Ziegemann O, Krumwiede L, Fessler E, Jae LT. Sensing, signaling and surviving mitochondrial stress. Cell Mol Life Sci. 2021;78:5925–51.
Guo X, et al. Mitochondrial stress is relayed to the cytosol by an OMA1–DELE1–HRI pathway. Nature. 2020;579:427–32.
Zhou D, et al. Phosphorylation of eIF2 directs ATF5 translational control in response to diverse stress conditions. J Biol Chem. 2008;283:7064–73.
Rath E, et al. Induction of dsRNA-activated protein kinase links mitochondrial unfolded protein response to the pathogenesis of intestinal inflammation. Gut. 2012;61:1269–78.
Zhao Q, et al. A mitochondrial specific stress response in mammalian cells. EMBO J. 2002;21:4411–9.
Fiorese CJ, et al. The transcription factor ATF5 mediates a mammalian mitochondrial UPR. Curr Biol. 2016;26:2037–43.
Yung HW, et al. Noncanonical mitochondrial unfolded protein response impairs placental oxidative phosphorylation in early-onset preeclampsia. Proc Natl Acad Sci USA. 2019;116:18109–18.
Wang YT, et al. RAPID REPORT signaling and stress response cardioprotection by the mitochondrial unfolded protein response requires ATF5. Am J Physiol Heart Circ Physiol. 2019;317:472–8.
Shen Y, et al. Activation of mitochondrial unfolded protein response in SHSY5Y expressing APP cells and APP/PS1 mice. Front Cell Neurosci. 2020;13:568.
Restelli LM, et al. Neuronal mitochondrial dysfunction activates the integrated stress response to induce fibroblast growth factor 21. Cell Rep. 2018;24:1407–14.
Stoica R, De Vos KJ, Paillusson S, Mueller S, Sancho RM, Lau KF, Vizcay-Barrena G, Lin WL, Xu YF, Lewis J, Dickson DW, Petrucelli L, Mitchell JC, Shaw CE, Miller CC. ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat Commun. 2014;5:3996. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncomms4996.
Stockwell SR, et al. Mechanism-based screen for G1/S checkpoint activators identifies a selective activator of EIF2AK3/PERK signalling. PLoS One. 2012;7(1):e28568.
Chen T, et al. Chemical genetics identify eIF2α kinase heme-regulated inhibitor as an anticancer target. Nat Chem Biol. 2011;7:610–6.
Boyce M, et al. A selective inhibitor of elF2α dephosphorylation protects cells from ER stress. Science. 2005;1979(307):935–9.
Tsaytler P, Harding HP, Ron D, Bertolotti A. Selective inhibition of a regulatory subunit of protein phosphatase 1 restores proteostasis. Science. 2011;1979(332):91–4.
Holmes B, Brogden RN, Heel RC, Speight TM, Avery GS. Guanabenz A review of its pharmacodynamic properties and therapeutic efficacy in hypertension. Drugs. 1983;26:212–29.
Das I, et al. Preventing proteostasis diseases by selective inhibition of a phosphatase regulatory subunit. Science. 2015;1979(348):239–42.
Sidrauski C. et al. Pharmacological brake-release of mRNA translation enhances cognitive memory. Elife 2013;2.
Sekine Y, et al. Mutations in a translation initiation factor identify the target of a memory-enhancing compound. Science. 2015;1979(348):1027–30.
Tsai JC. et al. Structure of the nucleotide exchange factor eIF2B reveals mechanism of memory-enhancing molecule. Science (1979) 2018;359.
Zyryanova AF, et al. Binding of ISRIB reveals a regulatory site in the nucleotide exchange factor eIF2B. Science. 2018;1979(359):1533–6.
Zyryanova AF, et al. ISRIB blunts the integrated stress response by allosterically antagonising the inhibitory effect of phosphorylated eIF2 on eIF2B. Mol Cell. 2021;81:88-103.e6.
Rabouw HH, et al. Small molecule ISRIB suppresses the integrated stress response within a defined window of activation. Proc Natl Acad Sci U S A. 2019;116:2097–102.
Axten JM, et al. Discovery of 7-Methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1 H -indol-5-yl)-7 H -pyrrolo[2,3- d ]pyrimidin-4-amine (GSK2606414), a Potent and Selective First-in-Class Inhibitor of Protein Kinase R (PKR)-like Endoplasmic Reticulum Kinase (PERK). J Med Chem. 2012;55:7193–207.
Axten JM, et al. Discovery of GSK2656157: An optimized PERK inhibitor selected for preclinical development. ACS Med Chem Lett. 2013;4:964–8.
Jammi NV, Whitby LR, Beal PA. Small molecule inhibitors of the RNA-dependent protein kinase. Biochem Biophys Res Commun. 2003;308:50–7.
Szaruga M. et al. Activation of the integrated stress response by inhibitors of its kinases. Nat Commun 2023;14.
Bellato HM, Hajj GNM. Translational control by eIF2α in neurons: Beyond the stress response. Cytoskeleton. 2016;73:551–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cm.21294.
Cagnetta R, et al. Noncanonical modulation of the eIF2 pathway controls an increase in local translation during neural wiring. Mol Cell. 2019;73:474-489.e5.
Kelleher RJ, Bear MF. The autistic neuron: troubled translation? Cell. 2008;135:401–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2008.10.017.
Costa-Mattioli M, et al. Translational control of hippocampal synaptic plasticity and memory by the eIF2α kinase GCN2. Nature. 2005;436:1166–70.
Hernandez PJ, Abel T. The role of protein synthesis in memory consolidation: progress amid decades of debate. Neurobiol Learn Mem. 2008;89:293–311.
Sutton MA, Schuman EM. Dendritic protein synthesis, synaptic plasticity, and memory. Cell. 2006;127:49–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2006.09.014.
Holt CE, Martin KC, Schuman EM. Local translation in neurons: visualization and function. Nat Struct Mol Biol. 2019;26:557–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41594-019-0263-5.
Lockshin ER, Calakos N. The integrated stress response in brain diseases: A double-edged sword for proteostasis and synapses. Curr Opin Neurobiol. 2024;87. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.conb.2024.102886.
Costa-Mattioli M, et al. eIF2α phosphorylation bidirectionally regulates the switch from short- to long-term synaptic plasticity and memory. Cell. 2007;129:195–206.
Sossin WS, Costa-Mattioli M. Translational control in the brain in health and disease. Cold Spring Harb Perspect Biol 2019;11.
Oliveira MM, Klann E. eIF2-dependent translation initiation: Memory consolidation and disruption in Alzheimer’s disease. Semin Cell Dev Biol. 2022;125:101–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2021.07.009.
Chen A. et al. Inducible enhancement of memory storage and synaptic plasticity in transgenic mice expressing an inhibitor of ATF4 (CREB-2) and C/EBP proteins sion of short-to long-term memory and short-to long-term synaptic plasticity in both mice (Abel et al Recent Studies Revealed That, in Addition To. Neuron. 2003;39.
Zhu S, Henninger K, McGrath BC, Cavener DR. PERK Regulates working memory and protein synthesis-dependent memory flexibility. PLoS One. 2016;11(9):e0162766.
Pasini S, Corona C, Liu J, Greene LA, Shelanski ML. Specific downregulation of hippocampal ATF4 reveals a necessary role in synaptic plasticity and memory. Cell Rep. 2015;11:183–91.
Liu J, Pasini S, Shelanski ML, Greene LA. Activating transcription factor 4 (ATF4) modulates post-synaptic development and dendritic spine morphology. Front Cell Neurosci. 2014;8:177.
Sharma V, et al. Local inhibition of PERK enhances memory and reverses age-related deterioration of cognitive and neuronal properties. J Neurosci. 2018;38:648–58.
Sapkota D, et al. Activity-dependent translation dynamically alters the proteome of the perisynaptic astrocyte process. Cell Rep. 2022;41(3):111474.
Sharm V, et al. mRNA translation in astrocytes controls hippocampal long-term synaptic plasticity and memory. Proc Natl Acad Sci U S A. 2023;120(49):e2308671120.
Lee HG, Lee JH, Flausino LE, Quintana FJ. Neuroinflammation: An astrocyte perspective. Sci Transl Med. 2023;15(721):eadi7828.
Barreda-Manso MA, Yanguas-Casás N, Nieto-Sampedro M, Romero-Ramírez L. Neuroprotection and blood-brain barrier restoration by salubrinal after a cortical stab injury. J Cell Physiol. 2017;232:1501–10.
Anuncibay-Soto B, et al. Post-ischemic salubrinal treatment results in a neuroprotective role in global cerebral ischemia. J Neurochem. 2016;138:295–306.
Wilson EH, Weninger W, Hunter CA. Trafficking of immune cells in the central nervous system. J Clin Invest. 2010;120:1368–79. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI41911.
Ueno M, et al. The expression of matrix metalloproteinase-13 is increased in vessels with blood-brain barrier impairment in a stroke-prone hypertensive model. Hypertens Res. 2009;32:332–8.
Cai XH, et al. Endoplasmic reticulum stress plays critical role in brain damage after chronic intermittent hypoxia in growing rats. Exp Neurol. 2014;257:148–56.
Celardo I, et al. Mitofusin-mediated ER stress triggers neurodegeneration in pink1/parkin models of Parkinson’s disease. Cell Death Dis. 2016;7(6):e2271.
Ryoo HD. The integrated stress response in metabolic adaptation. J Biol Chem. 2024;300(4):107151. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jbc.2024.107151.
Longchamp A, et al. Amino acid restriction triggers angiogenesis via GCN2/ATF4 regulation of VEGF and H2S production. Cell. 2018;173:117-129.e14.
Wek RC, Jackson BM, Hinnebusch AG. Juxtaposition of domains homologous to protein kinases and Histidyl-TRNA synthetases in GCN2 protein suggests a mechanism for coupling GCN4 expression to amino acid availability (Translational Control/Aminoacyl-TRNA Synthetase). Proc Nail Acad Sci USA. 1989;86.
Trinh MA, Klann E. Translational control by eIF2α kinases in long-lasting synaptic plasticity and long-term memory. Neurobiol Learn Mem. 2013;105:93–9.
Ohno M. Roles of eIF2α kinases in the pathogenesis of Alzheimer’s disease. Front Mol Neurosci. 2014;7:22. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnmol.2014.00022.
Ishimura R, Nagy G, Dotu I, Chuang JH, Ackerman SL. Activation of GCN2 kinase by ribosome stalling links translation elongation with translation initiation. Elife 2016.https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.14295.001
Wu CCC, Peterson A, Zinshteyn B, Regot S, Green R. Ribosome collisions trigger general stress responses to regulate cell fate. Cell. 2020;182:404-416.e14.
Devi L, Ohno M. Deletion of the eIF2α kinase GCN2 fails to rescue the memory decline associated with Alzheimer’s disease. PLoS One. 2013;8(10):e77335.
Kedersha N, et al. Evidence that ternary complex (eIF2-GTP-tRNA i Met)-deficient preinitiation complexes are core constituents of mammalian stress granules. Mol Biol Cell. 2002;13:195–210.
Kedersha NL, Gupta M, Li W, Miller I, Anderson P. RNA-binding proteins TIA-1 and TIAR link the phosphorylation of EIF-2 to the assembly of mammalian stress granules. J Cell Biol. 1999;147. http://www.jcb.org.
Mukherjee M, et al. ER stress impedes regulated secretion by governing key exocytotic and granulogenic molecular switches. 2023;(6):jcs261257. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2023.04.18.537291.
Allanach JR, Farrell JW, Mésidor M, Karimi-Abdolrezaee S. Current status of neuroprotective and neuroregenerative strategies in multiple sclerosis: a systematic review. Mult Scler J. 2022;28:29–48.
Murúa SR, Farez MF, Quintana FJ. The Immune Response in Multiple Sclerosis. Annu Rev Pathol Mech Dis. 2022;17:2021.
Ní Mháille A, et al. Increased expression of endoplasmic reticulum stress-related signaling pathway molecules in multiple sclerosis lesions. J Neuropathol Exp Neurol. 2008;67:200–11.
Ní Fhlathartaigh M. et al. Calreticulin and other components of endoplasmic reticulum stress in rat and human inflammatory demyelination. Acta Neuropathol Commun 2013;2.
Lin W, Harding HP, Ron D, Popko B. Endoplasmic reticulum stress modulates the response of myelinating oligodendrocytes to the immune cytokine interferon-γ. J Cell Biol. 2005;169:603-12.
Lin W, et al. The integrated stress response prevents demyelination by protecting oligodendrocytes against immune-mediated damage. J Clin Investig. 2007;117:448–56.
Lin W, et al. Enhanced integrated stress response promotes myelinating oligodendrocyte survival in response to interferon-γ. Am J Pathol. 2008;173:1508–17.
Hussien Y, Cavener DR, Popko B. Genetic inactivation of PERK signaling in mouse oligodendrocytes: Normal developmental myelination with increased susceptibility to inflammatory demyelination. Glia. 2014;62:680–91.
Chen Y, et al. Sephin1, which prolongs the integrated stress response, is a promising therapeutic for multiple sclerosis. Brain. 2019;142:344–61.
Chen Y, Kunjamma RB, Weiner M, Chan JR, Popko B. Prolonging the integrated stress response enhances cns remyelination in an inflammatory environment. Elife 2021;10.
Yue Y, Stanojlovic M, Lin Y, Karsenty G, Lin W. Oligodendrocyte-specific ATF4 inactivation does not influence the development of EAE. J Neuroinflamm. 2019;16:23.
Chen Y, et al. Insights into the mechanism of oligodendrocyte protection and remyelination enhancement by the integrated stress response. Glia. 2023;2180-2195. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/glia.24386.
Pernin F, et al. Regulation of stress granule formation in human oligodendrocytes. Nat Commun. 2024;15(1):1524.
Lin W, et al. Oligodendrocyte-specific activation of PERK signaling protects mice against experimental autoimmune encephalomyelitis. J Neurosci. 2013;33:5980–91.
Campbell GR, Ziabreva I, Reeve AK, Krishnan KJ, Reynolds R, Howell O. Mitochondrial DNA deletions and neurodegeneration in multiple sclerosis. Ann Neurol. 2011;69:481–92.
Mahad DH, Trapp BD, Lassmann H. Pathological mechanisms in progressive multiple sclerosis. Lancet Neurol. 2015;14:183–93.
Steudler J, Ecott T, Ivan DC, Bouillet E, Walthert S, Berve K. Autoimmune neuroinflammation triggers mitochondrial oxidation in oligodendrocytes. Glia. 2022;70:2045–61.
Abubakar MB et al. Alzheimer’s disease: an update and insights into pathophysiology. Front Aging Neurosci. 2022;14. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnagi.2022.742408.
Salminen A, Kauppinen A, Suuronen T, Kaarniranta K, Ojala J. ER stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology. J Neuroinflamm. 2009;6. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1742-2094-6-41.
Katayama T, et al. Induction of neuronal death by ER stress in Alzheimer’s disease. J Chem Neuroanatomy. 2004;28:67–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jchemneu.2003.12.004.
Ho Y-S, et al. Endoplasmic reticulum stress induces tau pathology and forms a vicious cycle: implication in Alzheimer’s disease pathogenesis. J Alzheimers Dis. 2012;28(4):839-54.
Radford H, Moreno JA, Verity N, Halliday M, Mallucci GR. PERK inhibition prevents tau-mediated neurodegeneration in a mouse model of frontotemporal dementia. Acta Neuropathol. 2015;130:633–42.
Lee DY, et al. Activation of PERK signaling attenuates Aβ-mediated ER stress. PLoS One. 2010;(5):e10489.
Bruch J, et al. PERK activation mitigates tau pathology in vitro and in vivo. EMBO Mol Med. 2017;9:371–84.
Wong TH, et al. EIF2AK3 variants in Dutch patients with Alzheimer’s disease. Neurobiol Aging. 2019;73(229):e11-229.e18.
Park G. et al. Neurodegeneration risk factor, EIF2AK3 (PERK), influences tau protein aggregation. J Biol Chem 2023;299.
Chuen R, et al. Involvement of double-stranded RNA-dependent protein kinase and phosphorylation of eukaryotic initiation factor-2a in neuronal degeneration. J Neurochem. 2002;83:1215-25.
Reimer L, et al. PKR kinase directly regulates tau expression and Alzheimer’s disease-related tau phosphorylation. Brain Pathol. 2021;31:103–19.
Sorrentino V, et al. Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature. 2017;552:187–93.
Beck JS, Mufson EJ, Counts SE, Alzheimer C. Author R. Evidence for Mitochondrial UPR Gene Activation in Familial and Sporadic Alzheimer’s Disease HHS Public Access Author Manuscript. Curr Alzheimer Res 2016;13.
Baleriola J, et al. Axonally synthesized ATF4 transmits a neurodegenerative signal across brain regions. Cell. 2014;158:1159–72.
Hosoi T, Kakimoto M, Tanaka K, Nomura J, Ozawa K. Unique pharmacological property of ISRIB in inhibition of Aβ-induced neuronal cell death. J Pharmacol Sci. 2016;131:292–5.
Hu Z. et al. Inhibition of the ISR abrogates mGluR5-dependent long-term depression and spatial memory deficits in a rat model of Alzheimer’s disease. Transl Psychiatry 2022;12.
Menšíková K. et al. Lewy body disease or diseases with Lewy bodies? npj Parkinson’s Dis. 2022;8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41531-021-00273-9.
Jankovic J, Tan EK. Parkinson’s disease: Etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020;91:795–808.
Pickles S, Vigié P, Youle RJ. Mitophagy and quality control mechanisms in mitochondrial maintenance. Curr Biol. 2018;28:R170–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cub.2018.01.004.
Chakrabarty Y, Yang Z, Chen H, Chan DC. The HRI branch of the integrated stress response selectively triggers mitophagy. Mol Cell. 2024;84:1090-1100.e6.
Mercado G, et al. Targeting PERK signaling with the small molecule GSK2606414 prevents neurodegeneration in a model of Parkinson’s disease. Neurobiol Dis. 2018;112:136–48.
Hoozemans JJM, et al. Activation of the unfolded protein response in Parkinson’s disease. Biochem Biophys Res Commun. 2007;354:707–11.
Bellucci A, et al. Induction of the unfolded protein response by α-synuclein in experimental models of Parkinson’s disease. J Neurochem. 2011;116:588–605.
Bouman L, et al. Parkin is transcriptionally regulated by ATF4: Evidence for an interconnection between mitochondrial stress and ER stress. Cell Death Differ. 2011;18:769–82.
Sun X, et al. ATF4 protects against neuronal death in cellular Parkinson’s disease models by maintaining levels of parkin. J Neurosci. 2013;33:2398–407.
Aimé P, et al. Trib3 is elevated in Parkinson’s disease and mediates death in Parkinson’s disease models. J Neurosci. 2015;35:10731–49.
Gully JC, et al. Up-regulation of activating transcription factor 4 induces severe loss of dopamine nigral neurons in a rat model of Parkinson’s disease. Neurosci Lett. 2016;627:36–41.
Imai Y, et al. An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress. Is a Substrate of Parkin Cell. 2001;105:891–902.
Chinta SJ, Mallajosyula JK, Rane A, Andersen JK. Mitochondrial α-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo. Neurosci Lett. 2010;486:235–9.
Luth ES, Stavrovskaya IG, Bartels T, Kristal BS, Selkoe DJ. Soluble, prefibrillar α-synuclein oligomers promote complex I-dependent, Ca2+-induced mitochondrial dysfunction. J Biol Chem. 2014;289:21490–507.
Wang W, et al. Parkinson’s disease-associated mutant VPS35 causes mitochondrial dysfunction by recycling DLP1 complexes. Nat Med. 2016;22:54–63.
Ibanez L. et al. Parkinson disease polygenic risk score is associated with Parkinson disease status and age at onset but not with alpha-synuclein cerebrospinal fluid levels. BMC Neurol 2017;17.
Demmings MD, Tennyson EC, Petroff GN, Tarnowski-Garner HE, Cregan SP. Activating transcription factor-4 promotes neuronal death induced by Parkinson’s disease neurotoxins and α-synuclein aggregates. Cell Death Differ. 2021;28:1627–43.
Masrori P, Van Damme P. Amyotrophic lateral sclerosis: a clinical review. Eur J Neurol. 2020;27:1918–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/ene.14393.
Mead RJ, Shan N, Reiser HJ, Marshall F, Shaw PJ. Amyotrophic lateral sclerosis: a neurodegenerative disorder poised for successful therapeutic translation. Nat Rev Drug Discov. 2023;22:185–212. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41573-022-00612-2.
Jo M, et al. The role of TDP-43 propagation in neurodegenerative diseases: integrating insights from clinical and experimental studies. Exper Mol Med. 2020;52:1652–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s12276-020-00513-7.
Ilieva EV, et al. Oxidative and endoplasmic reticulum stress interplay in sporadic amyotrophic lateral sclerosis. Brain. 2007;130:3111–23.
Nishitoh H, et al. ALS-linked mutant SOD1 induces ER stress- and ASK1-dependent motor neuron death by targeting Derlin-1. Genes Dev. 2008;22:1451–64.
Prell T, Lautenschläger J, Witte OW, Carri MT, Grosskreutz J. The unfolded protein response in models of human mutant G93A amyotrophic lateral sclerosis. Eur J Neurosci. 2012;35:652–60.
Chen D, Wang Y, Chin ER. Activation of the endoplasmic reticulum stress response in skeletal muscle of G93a*SOD1 amyotrophic lateral sclerosis mice. Front Cell Neurosci 2015;9.
Sun S, et al. Translational profiling identifies a cascade of damage initiated in motor neurons and spreading to glia in mutant sod1-mediated ALS. Proc Natl Acad Sci U S A. 2015;112:E6993–7002.
Kramer NJ, et al. CRISPR-Cas9 screens in human cells and primary neurons identify modifiers of C9ORF72 dipeptide-repeat-protein toxicity. Nat Genet. 2018;50:603–12.
Zu T, et al. Metformin inhibits RAN translation through PKR pathway and mitigates disease in C9orf72 ALS/ FTD mice. Proc Natl Acad Sci U S A. 2020;117:18591–9.
Kim HJ, et al. Therapeutic modulation of eIF2α phosphorylation rescues TDP-43 toxicity in amyotrophic lateral sclerosis disease models. Nat Genet. 2014;46:152–60.
Wang L, Popko B, Roos RP. An enhanced integrated stress response ameliorates mutant SOD1-induced ALS. Hum Mol Genet. 2014;23:2629–38.
Saxena S, Cabuy E, Caroni P. A role for motoneuron subtype-selective ER stress in disease manifestations of FALS mice. Nat Neurosci. 2009;12:627–36.
Luan W, et al. Early activation of cellular stress and death pathways caused by cytoplasmic TDP-43 in the rNLS8 mouse model of ALS and FTD. Mol Psychiatry. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41380-023-02036-9.
Szewczyk B. et al. FUS ALS neurons activate major stress pathways and reduce translation as an early protective mechanism against neurodegeneration. Cell Rep 2023;42.
Krols M, et al. Mitochondria-associated membranes as hubs for neurodegeneration. Acta Neuropathol. 2016;131:505–23.
Dafinca R, Barbagallo P, Talbot K. The role of mitochondrial dysfunction and ER stress in TDP-43 and C9ORF72 ALS. Front Cell Neurosci. 2021;15:653688. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fncel.2021.653688.
McCann EP, et al. Evidence for polygenic and oligogenic basis of Australian sporadic amyotrophic lateral sclerosis. J Med Genet. 2021;58:87–95.
Jiang HQ, et al. Guanabenz delays the onset of disease symptoms, extends lifespan, improves motor performance and attenuates motor neuron loss in the SOD1 G93A mouse model of amyotrophic lateral sclerosis. Neuroscience. 2014;277:132–8.
Dalla Bella E, et al. The unfolded protein response in amyotrophic later sclerosis: Results of a phase 2 trial. Brain. 2021;144:2635–47.
Sun L, et al. The integrated stress response is modulated by eIF2B agonist DNL343: results from phase 1 healthy subject and phase 1b ALS patient studies (P8–8.010). Neurology. 2023;100:3555.
Acknowledgements
S.K-A acknowledges the support by the Canadian Institute of Health Research and the Multiple Sclerosis Canada. MABJ was supported by a Studentship from Research Manitoba and Children’s Hospital Research Institute of Manitoba and received Hillary Kaufman Memorial Funds in Multiple Sclerosis from the University of Manitoba.
Author information
Authors and Affiliations
Contributions
MAB-J, SS and SK-A designed and wrote the manuscript. All authors have approved the final version of the manuscript.
Corresponding author
Ethics declarations
Competing interests
Authors declare no competing financial interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Bravo-Jimenez, M.A., Sharma, S. & Karimi-Abdolrezaee, S. The integrated stress response in neurodegenerative diseases. Mol Neurodegeneration 20, 20 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13024-025-00811-6
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13024-025-00811-6