Abstract
Most cellular protein synthesis, including synthesis of membrane-targeted and secreted proteins, which are critical for cellular and organ crosstalk, takes place at the endoplasmic reticulum (ER), placing the ER at the nexus of cellular signaling, growth, metabolism, and stress sensing. Ample evidence has established the dysregulation of protein homeostasis and the ER unfolded protein response (UPR) in cardiovascular disease. However, the mechanisms of stress sensing and signaling in the ER are incompletely defined. Recent studies have defined notable functions for the inositol-requiring kinase 1 (IRE1)/X-box- binding protein-1 (XBP1) branch of the UPR in regulation of cardiac function. This review highlights the mechanisms underlying IRE1 activation and the IRE1 interactome, which reveals unexpected functions for the UPR and summarizes our current understanding of the functions of IRE1 in cardiovascular disease.
Introduction
Protein synthesis and folding are indispensable for physiological cellular function. During protein synthesis, ribosomes direct nascent polypeptides to the endoplasmic reticulum (ER). This transfer takes place upon translation of a short peptide (16–30 amino acids long) signal peptide or targeting peptide at the N-terminus (or occasionally C-terminus) of the newly synthesized protein. The signal peptide is present in proteins that reside in the lumen of the organelles that make up the secretory pathway (endomembrane system), as well as in proteins secreted into the extracellular space, and inserted into most cellular membranes [1]. The ER represents a highly specialized cellular compartment, which, due to its oxidizing environment, is especially well-suited for expression and oxidative folding of the ER-resident proteins, proteins of the endomembrane system, and secreted proteins [1]. The process of controlling polypeptide influx into the ER, posttranslational modification, protein folding, protein export, and degradation of misfolded proteins is controlled tightly by the highly coordinated complex regulation of protein homeostasis or proteostasis.
The endoplasmic reticulum unfolded protein response (UPR) plays a central role in proteostasis, and is activated during physiological (cell growth, mitosis) and pathological stress stimuli (heat, hypoxia, and ischemia), to combat increased misfolded protein burden or declining folding capacity. The mammalian UPR, as known to date, consists of three main signaling protein branches, mediated by three ER transmembrane receptors: inositol-requiring kinase 1 (IRE1), protein kinase R-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6).
When subjected to ER stressors, cells react within as early as 15–30 min to halt their translational machinery via PERK activation and phosphorylation of its substrate, the eukaryotic translation initiation factor-2α [2,3]. IRE1 activation (see the following chapter) occurs in the same timeframe [4]. Around 4 h after exposure, downstream transcription factors, such as the activating transcription factor 4 (ATF4), which acts downstream of PERK, are actively translated and translocated into the nucleus. ATF6 is an ER-membrane resident protein that is transported to the Golgi apparatus and cleaved into its active form starting at ~1 h after the onset of ER stress, although earlier activation has been reported upon disruption of disulfide bonds of ER proteins caused by the reducing agent dithiothreitol [4]. The transcriptional targets of ATF6, mainly chaperones that aid in increasing protein-folding capacity in the ER, emerge at ~6 h after ER stress stimulus [5]. This initial burst of signaling from all three UPR branches is regarded as the protective, adaptive phase of UPR. If ER stress persists, the ATF6 and IRE1 pathways are turned off, despite continuous stimulation of UPR signaling, while the PERK pathway remains active [6], thereby switching to the maladaptive phase, where cell death programs are activated, preventing further damage to the organism (Figure 1a).
Figure 1.

IRE1 activation and mechanisms of action. (a) Graphical representation of activation and deactivation timeline of each of the UPR signaling pathways and dynamic crosstalk between the pathways. Thickness of the bar indicates pathway activity. Time is approximated as hours after ER stress induction, which is dependent on the model and strength of stress stimulus. IRE1 and PERK pathways generally antagonize each other through p58IPK and PRAR2 [100,101]. (b) Overview of the IRE1 activation process and IRE1 interactions with subcellular structures. (c) Illustration of known effector pathways of IRE1: (i) XBP1 is spliced by IRE1 in response to ER stress to produce spliced XBP1 (XBP1s), phosphorylated by p38 mitogen-activated protein kinase [102], and translocated to the nucleus, facilitated by p85α/β [103]. In the nucleus, XBP1 binds to cis-acting elements consisting of ER stress-response element (ERSE), ERSE-II, and unfolded response-pathway element (URPE) [104]. (ii) IRE1 endonuclease activity performs sequence-specific (as noted in red) RIDD and RIDDLE to regulate stress response. (iii) Depiction of JNK pathway, NF-κB pathway, and (iv) ERK pathway downstream of IRE1.
Disruption of proteostasis is evident in many human diseases [7–9]. Understanding the molecular mechanisms of the UPR may benefit the diagnosis and therapeutic treatment of disease. Since its discovery, studies utilizing various in vitro and in vivo models of disease stress have led to an increasing understanding of the complex signaling pathways of the UPR [10]. As a result, chemical compounds enhancing proteostasis have been developed and tested, and exciting preclinical results have led to promising clinical trials [11] and even to the introduction of some of the compounds to the clinical setting [12]. Challenges remain to resolve the problem of targeting such a ubiquitous and integrative cellular network precisely in specific tissues and organs [13]. Moreover, modifying the UPR is potentially a double-edged sword. While it is an adaptive biochemical process that regulates cell homeostasis and maintenance of normal physiological function, prolonged ER stress can push the UPR past beneficial functions, such as reduced protein production and increased folding and clearance, to apoptotic signaling [14]. Furthermore, recent studies demonstrate cell-type-specific aspects of the UPR, which are mainly driven by tissue-specific differences in the differential demand on ER capacity and function [15]. Therefore, detailed understanding of the UPR in specific cell types and disease models is critical for adopting treatment options in a clinical setting. The IRE1 pathway is considered the most evolutionarily conserved branch, orchestrating the UPR in yeast [16,17] and most other eukaryotes such as plants, fungi, and most metazoans [18]. This is reflected in the plethora of its downstream signaling pathways, which are switched on and off, depending on the stress phase, reflecting opposing effects from cell survival to mediating apoptosis (Figure 1a and c). This review will focus on the details of the complexities of the IRE1 branch of the UPR, as well as on our current knowledge of the roles of the IRE1 branch in cardiovascular disease.
Inositol-requiring kinase 1 stress sensing and activation
While yeast IRE1 only has one isoform, the mammalian IRE1 consists of two isoforms: IRE1α and IRE1β. IRE1α, encoded by the ERN1 gene, is expressed ubiquitously, while IRE1β, encoded by the ERN2 gene, is expressed in lung and intestinal epithelial cells [19] (for simplicity, we will use the term ‘IRE1’ in the rest of this review to refer to the yeast IRE1 and the mammalian IRE1α). Three models of stress sensing have been proposed to lead to initiation of the IRE1 pathway. In the original (competitive) model, it was proposed that in resting state, IRE1 is associated with the ER-resident chaperone, binding-immunoglobulin protein also named 78 kDa glucose-regulated protein (BiP, GRP78). When high amounts of misfolded proteins are present in the ER, BiP preferentially binds to these proteins, thereby dissociating from IRE1 [20]. This release allows IRE1 to multimerize and autophosphorylate its cytosolic domain, triggering the activation of the IRE1 RNase activity, initiating XBP1 mRNA splicing and UPR responses.
In addition to this competitive model, BiP has been suggested to bind to IRE1 via its ATPase domain and undergo allosteric changes, thereby dissociating from IRE1 when unfolded proteins are bound to its substrate-binding domain during UPR [21]. Mutagenesis studies of the BiP-binding site on yeast IRE1 have shown that IRE1 retains its ER stress-sensing ability, despite deletion of the BiP-binding site [22]. Evidence of direct binding to unfolded proteins suggests that this is sufficient to activate yeast IRE1 in response to unfolded protein accumulation [23]. Similar results have been shown in mammalian cells [24–26], where IRE1 activation seems to be more complicated. Recent studies not only indicate that preformed oligomers of ER stress sensors, such as IRE1, exist in a resting state [24], but also hetero-oligomers consisting of IRE1 and other regulatory proteins modify IRE1 activity during resting conditions and stress exposure [27]. While extensive biochemical characterization of the IRE1 interactome is required to determine the dynamics of its protein composition under conditions of mild and chronic ER stress, studies to date suggest that IRE1 is involved in cellular signaling, protein synthesis, and targeting [28] (Table 1).
Table 1.
The list of proteins identified as IRE1-interacting partners.
| Interacting partner | ER state and cell type | Effect | Source |
|---|---|---|---|
| Activating interaction partners | |||
| TRAF2 | TG, TM; HEK293 | Direct binding partner activates JNK | [40] |
| JIK | Resting state, HEK293 | Direct binding partner of IRE1 and TRAF2 modulates IRE1–TRAF2 complex | [105] |
| ASK1 | TG, TM; PC-12 | Direct binding partner of TRAF2 and IRE1 required to activate JNK | [39] |
| HSP90 | Resting state: COS7, AR72J | Direct binding partner of the cytosolic domain of IRE1, stabilizing it by inhibiting proteasomal degradation | [106] |
| PTP-1B | TM, Azc; MEF | Knockout of PTP-1B impaired IRE1 signaling, reducing XBP1 splicing and JNK activation. No data on physical interaction | [107] |
| TNFR1 | TG, TM; 3T3, HEK293, and MEF | Direct binding partner of IRE1 and RIP promotes JNK activation | [108] |
| BAX, BAK | TM; BAX and BAK double-knockout mouse livers, double- knockout MEF | Direct binding partner of the cytosolic domain of IRE1 promotes IRE1 activation | [109] |
| IKK | TM, TG; HEK293, H1299, PC-3, and MCF-7 | Direct binding partner of IRE1 and TRAF2 upon ER stress. IRE1 is required to mediate ER stress- induced NF-κB activation | [110] |
| AIP1 | TM; AIP1-knockout mice | Direct binding partner enhances IRE1 dimerization | [111] |
| HSP72 | TM, TG; PC-12 | Direct binding partner boosts XBP1 splicing | [112] |
| BIM, PUMA | TM, MEF | Direct binding partner facilitates XBP1 splicing | [113] |
| NMHCIIB | TG, TM; MEF, C. elegans | Direct binding partner promotes oligomerization and activates IRE1 cluster (foci) formation | [114] |
| PARP16 | TM, TG; HeLa | Direct binding partner required to activate IRE1–XBP1 pathway | [115] |
| RNF13 | TM; SHSY-5Y, STS-108 | Direct binding partner promotes its phosphorylation, XBP1 splicing, and JNK activation | [116] |
| Sig-1R | TG, heat shock; HeLa, AR42J, Neuro2A, and Du145 | Transient association with monomeric luminal domain of IRE1 in MAMs in very early phase of ER stress to promote IRE1–XBP1 pathway | [117] |
| Cab45S | TM, HeLa | Direct binding partner of BiP suppresses IRE1/JNK pathway by stabilizing BiP–RE1 interaction | [118] |
| CHIP | GA, HEK293 | Direct binding partner ubiquitinates IRE1, inducing IRE/TRAF2/JNK pathway and inhibiting senescence, but not IRE1–XBP1 pathway | [119] |
| ABL tyrosine kinases | TM; NOD mouse, INS-1 cells | Direct binding partner of the cytosolic domain of IRE1 and promotes its activation | [120] |
| RNH1 | TM, HK-2 cell line | Direct binding partner of the cytosolic RNase domain of IRE1. Decrease of RNH1 increases splicing of XBP1 | [121] |
| HSP47 | TM, MEF | Direct binding partner displaces BiP from IRE1 and promotes IRE1 oligomerization | [122] |
| Ribosome-associated complex | TG, DTT, celastrol; HeLa, HEK293 | Reduction of RAC inhibits IRE1 higher- order oligomerization | [123] |
| PRKCSH | TM, L02 normal liver cells | Direct binding partner promotes IRE1 autophosphorylation and oligomerization | [124] |
| Inhibitory interaction partners | |||
| Ptc2p phosphatase | TM, S. cerevisiae | Direct binding partner dephosphorylates IRE1 | [125] |
| Nck | Azc, MEF | Associates with IRE1 in vitro, inhibiting IRE1-mediated ERK-1 activation | [41] |
| JAB1 | Resting state, TM, TG; yeast, HEK293 | Direct binding partner inhibits IRE1 activity in resting state | [126] |
| Drc2 phosphatase | TM, S. cerevisiae | Direct binding partner dephosphorylates IRE1 and inhibits XBP1 splicing | [127] |
| Hrd1 | Resting state, HEK293 | Direct binding partner induces IRE1 ubiquitination and degradation | [128] |
| BI-1 | TM; BAX inhibitor-1 (BI-1)-knockout mice and MEF, D. melanogaster | Direct binding partner suppresses IRE1 RNAse activity | [129] |
| USP14 | TG; MEF, HEK293 | Direct binding partner of the cytosolic domain of IRE1 inhibits ER-associated protein degradation, dissociates from IRE1 upon ER stress | [130] |
| RACK1 | TM, TG; INS-1, HEK293 | Forms a complex with IRE1 and PP2A, promotes dephosphorylation of IRE1 by PP2A | [131] |
| PPM1l phosphatase | Resting state; HEK293, PPM1l- knockout mice | Direct binding partner dephosphorylates IRE1. Knockout mice display reduced IRE1 mRNA and increased JNK and caspase-12 activation | [132,133] |
| PDIA6 | TM, TG; 3T3, C. elegans | Direct binding partner of the luminal domain of IRE1 limits the duration of IRE1 activation. Loss of PDIA6 in C. elegans leads to ER stress hyperactivation | [134] |
| NMI | IL-1β + IFNγ; HEK293, INS-1E, rat beta cells | Direct binding partner knockdown of NMI increases IRE1/JNK signaling and apoptosis | [135] |
| UBD | IL-1β + IFNγ; HEK293, human and rat beta cells | Direct binding partner knockdown of UBD enhances JNK phosphorylation in an IRE1-independent manner. Does not regulate endonuclease activity of IRE1 | [136] |
| Sec61 | TG, TM; HEK293, MEF | Part of the hetero-oligomer of IRE1 recruits XBP1 to IRE1 to undergo splicing, limiting higher-order oligomer formation upon ER stress | [137,138] |
| ERdj4/DNAJB9 | Resting state, CHO | Direct binding partner of the luminal domain of IRE1 recruits BiP through the stimulation of ATP hydrolysis to disrupt IRE1 dimers | [139] |
| Fortilin | TG, EGF-SubA; PC-3- knockout mice | Direct binding partner of the cytosolic domain of IRE1 inhibits IRE1 kinase and RNAse activity, protects mouse hepatocytes through apoptosis inhibition | [140] |
| Calsequestrin | Resting states; COS, rabbit hind-leg muscle, mice ventricular cardiomyocytes | Associates with IRE1 in vitro and in COS cells. In muscle, cells colocalize with IRE1 in the junctional SR. Prevents IRE1 dimerization in vitro. Unknown in vivo function | [141] |
| Sec63 | TM, TG; HEK293 | Activates BiP ATPase, promoting its binding to IRE1, deactivating IRE1 | [142] |
| IRE1β | TG, human intestinal epithelial cell lines, HEK293 | Associates with IRE1 in polymers and inhibits XBP1 splicing in a dominant-negative manner | [143] |
| EI24 | Resting state, TM; HeLa | Direct binding partner of the kinase domain of IRE1. Inhibits IRE1 activation in resting state. Upon ER stress, EI24 dissociates from IRE1 to target IP3R1 to prevent ER calcium depletion. | [144] |
| Binding partners with noncanonical functions | |||
| Filamin A | Resting state; HEK293, MEFs, Huh7, D. melanogaster, and D. rerio mice | Direct binding partner regulates cytoskeleton dynamics upstream of filamin A and independent of XBP1 splicing | [145] |
| Structural RNA: signal- recognition particle (SRP) RNA, ribosomal and transfer RNAs | Resting state: TM, HEK293 | IRE1 interacts with tRNAs, rRNAs, and ribosomes on the ER surface with yet unknown physiological function | [46] |
| InsP3R at MAMs | Resting state: MEF, HEK293 mice | Interacts with IRE1 at MAMs, effector of a noncanonical IRE1 effector arm in controlling mitochondrial calcium, energetics and apoptosis | [47] |
The list of proteins identified as IRE1-interacting partners. For each interacting partner, the ER state and cell type in which the protein was investigated as well as the effects of the interaction with IRE1 are given. ER stressor compounds: TM: tunicamycin; TG: thapsigargin; Azc: azetidine-2-carboxylic acid; GA: geldanamycin; DTT: dithiothreitol; IFN-γ: interferon gamma; EGF-SubA: epidermal growth factor fused to the proteolytic A subunit of a bacterial AB5 toxin.
Signal transduction by inositol-requiring kinase 1
Upon activation, IRE1 dimerizes, trans-autophosphosphorylates, and oligomerizes (Figure 1b) [29], forming the kinase-extension nuclease domain [30], which possesses site-specific RNase activity to cleave HAC1 (homologous to ATF/CREB 1; yeast)/XBP1 (mammalian cells) [31]. IRE1 transduces signals in part through the excision of a 26-nucleotide intron of XBP1, resulting in spliced XBP1 (XBP1s), encoding a highly active transcription factor to enforce UPR [32]. Translated unspliced XBP1 (XBP1u) targets its own mRNA–ribosome-nascent chain complex to the vicinity of IRE1 at the membrane of ER, facilitating its splicing [33]. IRE1 RNAse also targets a subset of ER-membrane-localized mRNAs as well as some noncoding RNAs, in a mechanism termed regulated IRE1-dependent mRNA decay (RIDD) [34]. RIDD is sequence-specific and degrades hundreds of RNA targets upon ER stress, alleviating the load of proteins entering the ER and taking part in cell-fate decisions upon unresolvable ER stress [35,36]. Target selection and mechanisms of recruitment of specific RNA targets to IRE1 remain nebulous. Recently, by integrating nascent and global RNA transcriptomes in an IRE1-knockout model, IRE1α-dependent decay of a set of mRNAs lacking the RIDD sequence was identified: this was termed “RIDD lacking endo-motif” (RIDDLE). While XBP1 and RIDD targets are targeted by IRE1 dimers, it appears that only higher-order polyphospho-oligomers can perform RIDDLE [37]. Increasing knowledge of IRE1 rheostatic RNAse target selection may uncover a unique master switch governing UPR and cell fate [38].
Besides its endonuclease effector activity, IRE1 associates with TNF receptor-associated factor 2 (TRAF2) and apoptosis signal-regulating kinase 1 (ASK1) in a hetero-oligomer manner, inducing ASK1 to activate the c-Jun N-terminal kinase pathway (JNK–c-Jun) with its pleiotropic effector properties [39,40]. Extracellular signal-regulated kinase activation has also been found to be IRE1-dependent during azetidine-2-carboxylic acid (nonprotein amino acid proline homolog used to induce ER stress) and tunicamycin- (inhibitor of protein glycosylation) induced ER stress (Figure 1c) [41].
The proximal part of the IRE1–XBP1 pathway is induced early and attenuates its canonical signaling during a relatively early phase of ER stress compared with ATF6 and PERK (Figure 1a) [6]. XBP1u mediates deactivation of the IRE1–XBP1 signaling by binding to spliced XBP1 mRNA. The XBP1u protein–XBP1s mRNA complex is then degraded by the proteasome through a degradation domain on XBP1u [42]. XBP1u also functions as a negative regulator of XBP1s and activated ATF6, by targeting these proteins for destruction by the proteasome via direct association [43]. Moreover, XBP1u directly binds and promotes the degradation of forkhead box protein O1 (FOXO1), thereby suppressing autophagy [44]. Aside from targeting binding partners for degradation, XBP1u has been shown to reduce shear stress-induced oxidative stress in endothelial cells by complexing with histone deacetylase 3 and AKT1, promoting AKT1 phosphorylation and activating the phosphoinositide-3-kinase/AKT pathway [45].
Nevertheless, non-XBP1s-mediated signaling has been the focus of recent studies, revealing previously unknown functions of the IRE1–XBP1 pathway. By employing photoactivatable ribonucleoside-specific UV cross-linking, close physical association between IRE1, with a novel subset of structural noncoding RNAs and proteins associated with these RNAs, was identified, revealing a link between IRE1, the ribosome machinery, and the signal-recognition particle ribonucleoprotein complex [46]. This new interaction may shed light on a novel surveillance function of IRE1 on global translation. Another surprising association has been found in mitochondria-associated ER membranes (MAMs), a specialized ER subdomain within close physical proximity to the mitochondria (Figure 1b), where IRE1 enhances mitochondrial calcium uptake, increases mitochondrial ATP levels, and energetics, especially in response to ER stress, independent of the RNAse activity of IRE1. Observations in IRE1-knockout mouse hepatocytes showed altered mitochondrial morphology and significant reduction of InsP3R at MAMs. Interestingly, these mice developed decreased global glucose tolerance, indicating a cell nonautonomous effect, in which phenotypes were observed outside of the IRE1-knockout cells, posing possible therapeutic consequences [47]. Possible involvement of nonautonomous effects of IRE1 and XBP1 has also been demonstrated in C. elegans, where enforced neuronal XBP1s expression was sufficient to induce intestinal UPR through neurohumoral means and prolonged lifespan by increasing resistance to ER stress in adulthood, where UPR is constitutively downregulated [48]. These novel discoveries take IRE1 beyond a simple binary signaling switch to an interesting target in senescence and its associated diseases.
The unfolded protein response in the heart
Adult human cardiomyocytes are highly differentiated specialized cells without the ability of meaningful cell division or regeneration [49], highlighting the importance of maintaining proteostasis and cell integrity in the myocardium. It is no surprise that the UPR has been shown to be involved in the initiation and progression of cardiovascular disease [50], which is the most prominent disease burden in the elderly worldwide [51]. During disease progression in the heart, activation of all three branches of the UPR is evident. However, distinct differences in signaling output, depending on the pathophysiological stimulus, separate the activation of the different pathways. ATF6 is regarded as generally protective through its ability to induce the adaptive phase of the UPR by boosting the folding capacity of the ER when misfolded protein accumulation occurs in the diseased heart. This is evident in cardiac ischemia/reperfusion (I/R) models where ATF6 is activated during ischemia and ATF6 was shown to protect from I/R damage by upregulating ER chaperones and antioxidant and alleviating toxic cytosolic calcium levels [52,53]. In physiological and pathological growth of the heart, activation of ATF6 is required to drive adaptive myocyte hypertrophy [54]. Strikingly, although different stresses, such as I/R, pressure overload, or exercise, can activate ATF6, the consequent transcriptional reprogramming seems to be a stress-specific response [54]. PERK activation is an early event observed in the setting of I/R and promotes survival, but prolonged PERK activation is detrimental to myocyte survival by activating apoptosis through the ATF4/C/EBP homologous protein (CHOP) axis [55]. Similarly, PERK is cardioprotective during the early phases of pressure overload. In contrast, chronic heart failure results in downstream CHOP activation and apoptosis [55]. In contrast to ATF6 and PERK, the known impact of IRE1/XBP1 pathway on cardiovascular diseases is highly tissue-specific. In addition, multiple novel noncanonical functions of XBP1u/XBP1s have been uncovered to be important in cardiovascular diseases, further enlarging the network of possible signaling by IRE1.
The inositol-requiring kinase 1–X-box- binding protein-1 axis in cardiac remodeling and heart failure
Cardiovascular disease increases the risk of developing heart failure with reduced ejection fraction (HFrEF). Heart failure defines a clinical state where cardiac output is insufficient to meet oxygen demand [56]. The progression to heart failure is associated with myocardial remodeling, an adaption of the myocardium, extracellular matrix, and myocardial vasculature to disease stress [57]. Myocardial fibrosis, an important part of cardiac remodeling, is associated with adverse outcome of heart-failure patients and as such is key for diagnosis and therapeutic potential in heart failure [58]. Fibrosis is mediated by dynamic cardiac fibroblast cell states, which underlie the heart’s fibrotic response to maintain structural integrity in the short term, and result in cardiac fibrosis and maladaptive remodeling in the long term [57]. Activation of the IRE1–XBP1 pathway guides ER biogenesis and enhances ER capacity by expanding ER size through transcriptional activity of XBP1 [59]. Moreover, XBP1 is required for the development of specialized secretory cell types such as antibody-secreting plasma cells and pancreatic acinar cells [59]. Given that protein secretion is a feature of cardiac fibroblasts, it is plausible that the IRE1 branch of the UPR serves important roles in the regulation of the dynamic cell states of cardiac fibroblasts and, therefore, fibrosis. In a recent study, inducible, cardiomyocyte-specific overexpression of IRE1 resulted in potent but transient IRE1 activation [60]. Nevertheless, in response to pressure overload, the transgenic hearts exhibited preserved function and reduced fibrotic area, increased adaptive UPR signaling, and blunted inflammatory and pathological gene expression. However, XBP1s has been shown to act downstream of calreticulin expression in mice to induce cardiac fibrosis by TGFβ1/Smad2/3 signaling pathway activation, which was reversed by tauroursodeoxycholic acid [61]. Moreover, activation of Sigma-1 receptor by fluvoxamine inhibited cardiac fibrosis, apparently through downregulation of the IRE1–XBP1 pathway [62].
Upregulation of the UPR is observed in HFrEF patients [63,64]. Unloading the failing heart by implantation of a left ventricular assist device improves cardiac function and ameliorates the UPR [65]. In a rat model of pressure overload-induced hypertrophy via abdominal aortic constriction, XBP1s expression was dynamically regulated, with increasing expression in the compensatory hypertrophy phase up to 4 weeks after induction of pressure overload, and a subsequent decrease during the heart failure phase. This regulation was mediated by microRNA-214 and the microRNA-30* family, which reduce the expression of XBP1 and its targets during the late heart-failure stage [66,67]. Analogous to in vitro models where the dynamics of IRE1 signaling is extensively studied, this may reflect a protective function of IRE1 signaling during a relatively early phase of UPR in heart disease, while in a later maladaptive phase, IRE1 signaling is switched off. Spliced XBP1 is associated with the expression of brain natriuretic peptide (BNP) in neonatal rat cardiomyocytes by binding to an AP1/CRE-like element in the BNP promoter region and increasing its activity [68]. Conversely, recent evidence in heart failure with preserved ejection fraction (HFpEF) patients and rodent models of HFpEF unveiled inhibition of the IRE1–XBP1 pathway [69,70]. In the most widely accepted rodent model of HFpEF, feeding mice a high-fat diet and Nω-nitro-L-arginine methyl ester (L-NAME) resulted in a model of metabolic syndrome and hypertension and led to increased left ventricular filling pressure and decreased longitudinal myocardial strain. In that study, increased filling pressure led to pulmonary congestion and hypertrophy of cardiomyocytes, which resulted in exercise intolerance. Moreover, the HFpEF mice displayed reduction in cardiac IRE1 phosphorylation and decreased levels of XBP1s, BiP, and CHOP, in contrast to a HFrEF model undergoing transverse aortic constriction. Most interestingly, while IRE1–XBP1 signaling was downregulated, the PERK and ATF6 pathways were not, suggesting a possible UPR-independent regulation of the IRE1 pathway. Overexpression of XBP1s was sufficient to partially rescue the phenotype. Mechanistically, the authors found that application of l-NAME induced systemic inflammation, upregulating the inducible nitric oxide synthase in the heart, and inducing S-nitrosylation of IRE1, preventing XBP1 splicing. XBP1 splicing was restored in iNOS-knockout mice that were subjected to the HFpEF feeding protocol, while the HFpEF phenotype was partially rescued [69]. These findings suggest multi-organ regulation of the IRE1–XBP1 pathway as a pathological driver of HFpEF.
The inositol-requiring kinase 1–X-box- binding protein-1 axis in atherosclerosis and myocardial ischemia/reperfusion
Atherosclerosis develops through endothelial injury and subendothelial infiltration of monocytes and lipoproteins through a leaky, dysfunctional endothelium [71]. These lipoproteins are oxidized by nitric oxide (NO), produced by macrophages, and subsequently internalized to give rise to foam cells, which undergo apoptosis, forming the fatty streaks. Following advanced immune response, smooth muscle cells migrate to the site of inflammation, proliferate, and synthesize the extracellular matrix, which stabilizes the plaque while narrowing the vessel lumen, forming the fibrous plaque [71,72]. Acute coronary syndrome and stroke are caused by disruption of a vulnerable atherosclerotic fibrous plaque. A vulnerable plaque is the result of chronic inflammation, which leads to decreased collagen production and increased expression of collagenases, decreasing the width of the plaque cap and rendering it unstable, therefore prone to rupture and thromboembolic events [71,73].
Evidence of upregulated UPR in human atherosclerosis has been gathered in autopsies involving patients who succumbed to acute coronary syndrome and noncardiac causes, as well as biopsies from patients with stable or unstable angina pectoris. Immunostaining revealed increased expression of ER chaperones GRP78 and GRP94 as well as CHOP in smooth muscle cells and macrophages in patients with more severe disease (unstable angina, unstable thin plaques, and ruptured plaques in the deceased patients), forming a possible connection to reactive UPR signaling upon disease progression [74]. In a ApoE-knockout atherosclerosis mouse model, XBP1 splicing was found highly upregulated in the aortic endothelium in areas with disturbed blood flow, but not in regions with laminar flow. Forced expression of XBP1s induced atherosclerosis in isografted straight vessel with laminar flow, linking XBP1 splicing to the pathogenesis of atherosclerosis. Mechanistically, XBP1 splicing was found to be required for sheer stress-induced endothelial proliferation, and induced endothelial apoptosis through suppression of vascular endothelial (VE)-cadherin expression, which maintains endothelial integrity [75].
XBP1 splicing is also crucial for vascular smooth muscle cell (vSMC) proliferation, and is partially needed for vSMC migration, resulting in reduced neointima formation in a XBP1-knockout mouse model. XBP1 acts through suppression of interestingly in part by a transforming growth factor (TGF)-β-mediated paracrine mechanism, and in part through microRNA-18a and microRNA-1274B, which target calponin h1 transcript for degradation. Moreover, XBP1s forms a complex with PI3K and Akt and activates the Akt pathway and thereby, vSMC migration [76]. While XBP1s is involved in vSMC calcification downstream of bone morphogenetic protein 2-induced Runx2 expression in human coronary artery smooth muscle cells [77], a novel function of the unspliced XBP1 (XBP1u) has been uncovered in vSMC calcification. XBP1u levels were found to be reduced through proteasomal degradation in calcified vSMCs, calcified aorta of adenine diet-fed mice, and human radial arteries with chronic kidney failure. Inhibition of XBP1u promoted chondrogenic markers and overexpression of XBP1u reduced calcification in rat vSMC and XBP1 SMC-specific knockout mice fed with adenine diet. Through interactome analysis, β-catenin, a member of Wnt signaling governing calcification, was found to specifically bind to XBP1u, following which, XBP1u sequesters β-catenin for proteasome-mediated degradation [78]. In macrophages, several pro-atherogenic chemokines involved in lipid-driven inflammation have been identified to be IRE1-regulated, including interleukin-1 beta (IL-1β) and C-C motif chemokine 2 (CCL2). IRE1 or XBP1 inhibition abolished lipid-driven IL-1β through suppression of lipid-induced release of mitochondrial reactive oxygen species and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome. Treating ApoE-knockout mice with the specific IRE1 inhibitors STF-083010 or 4μ8c decreased atherosclerosis in vivo and modified plaque composition with markedly reduced macrophages, posing a possible mechanism in reducing plaque progression [79].
In the I/R model in cultured cardiomyocytes and mouse hearts, XBP1 splicing was increased during the ischemic phase [80]. While data point to a pro-atherogenic role for XBP1s, inhibition of XBP1s in the I/R setting is detrimental to cardiomyocytes [80,81]. Mechanistically, XBP1s was found to mediate transcription of genes encoding enzymes of the hexosamine biosynthetic pathway, including the rate-limiting glutamine fructose-6-phosphate aminotransferase 1 (GFAT1), all of which facilitate O-linked β-N-acetylglucosamine protein modification, which is necessary for correct protein folding. Expression of XBP1s in transgenic mice for 2 weeks followed by I/R decreased infarct size and preserved cardiac function. Suppression of GFAT1 in vitro and in vivo partially restored the protective effect [81]. XBP1s, inducible by vascular endothelial growth factor, promotes angiogenesis through PI3K/AKT/GSK3β pathway [82]. Altogether, these findings suggest cytoprotective and pro-angiogenic functions for the IRE1–XBP1 axis in the setting of ischemia and I/R in most of the cell types studied to date. Depending on cellular identity and function, the activation of this axis may preserve or exacerbate cardiovascular function.
Endoplasmic reticulum stress in diabetic cardiomyopathy
The diabetic heart is characterized by heart failure independent of coronary artery disease, hypertension, or valvular disease in diabetic patients. Early stages of diabetic cardiomyopathy consist of subcellular changes of signaling, and structural abnormalities of cellular compartments, which remain asymptomatic to the patients. Early clinical presentations are concentric hypertrophy, diastolic dysfunction, and increased myocardial fibrosis. Ultimately, systolic impairment will develop [83]. Guideline-driven diagnostic procedures or targeted treatment options are currently not available, although the emergence and success of sodium-glucose cotransporter-2 inhibitors (STLG2i) in treating HFrEF and HFpEF sheds light on the interplay of diabetes and heart failure [84]. On the molecular level, factors contributing to the still-vaguely understood pathogenesis include chronic inflammation, generation of excessive reactive oxygen species, accumulation of advanced glycation end products, and activation of signaling such as TGF-β, leading to increased myofibroblast differentiation, disturbance of extracellular matrix homeostasis, and increased cross-linking of extracellular matrix proteins [85].
ER stress is a key feature of metabolic disorders. UPR and IRE1 have been extensively studied in glucose metabolism and metabolic syndrome [86]. XBP1 deficiency is linked to β-cell dysfunction, while the IRE1–JNK pathway mediates intracellular insulin signaling in a cell-specific manner. In obesity, nuclear translocation of XBP1s is markedly decreased, and expression of XBP1 in adipocytes improves glucose tolerance and insulin sensitivity [87]. More recently, IRE1/XBP1s activation was found to promote systemic adaptive remodeling in obesity [88]. Specifically, XBP1s-selective pharmacological IRE1 activator, IXA4, transiently activated protective IRE1/XBP1s signaling in liver without inducing RIDD or TRAF2/JNK signaling. IXA4 treatment improved systemic glucose metabolism and liver insulin action through IRE1-dependent remodeling of the hepatic transcriptome that reduced glucose production and steatosis. IXA4-stimulated IRE1 activation also enhanced pancreatic function. These findings indicate that transient activation of IRE1/XBP1s signaling promotes integrative multitissue benefits that mitigate obesity-driven metabolic dysfunction.
Less data exist on possible IRE1/XBP1-related mechanisms in the diabetic heart, although IRE1 shares common pathways with key signaling cascades contributing to diabetic cardiac remodeling. The NLRP3 inflammasome is also triggered in a type-2 diabetes rat model fed with high-fat diet and injected with streptozotocin, which is specifically cytotoxic to pancreatic β-cells, leading to reduced systolic and diastolic heart function and increased fibrosis. Gene silencing of NLRP3 reversed cardiac function and fibrotic remodeling [89]. Bone morphogenetic protein-7, STLG2i, rosuvastatin, which has an anti-inflammatory effect on some cardiovascular diseases, and the blood thinner ticagrelor have been demonstrated to alleviate cardiac dysfunction in different diabetic rodent models [90–93]. IRE1 has been shown to regulate NLRP3 inflammasome activity via upregulation of thioredoxin-interacting protein during ER stress, and suppression of IRE1 via small-molecule RNase inhibitor counteracts disease progression in a diabetic mouse model [94]. However, direct evidence of possible disease-modulating effect of cardiac-specific IRE1 and possible pro-inflammatory effects in diabetic cardiomyopathy are still missing.
Summary and outlook
A plethora of studies have established IRE1 as a key regulator of cellular stress over the 30 years since its discovery by Kazutoshi Mori and Peter Walter, who independently, and according to Peter Walter, serendipitously, identified IRE1 in yeast [95,96].
Over the years, basic research has contributed to understanding stress sensing and activation of IRE1 through three models, competitive or allosteric dissociation of BiP, and direct substrate binding to IRE1 itself. It is feasible that these mechanisms are not exclusive, but mutually exist alongside each other. Understanding the contributions of these activating mechanisms in animal models and in relation to disease stress remains to be investigated. Furthermore, by employing emerging global profiling approaches in a forward-genetics setting, the extensive networks of effector signaling of the RNAse function of IRE1, through XBP1 splicing, RIDD, and RIDDLE, have revealed the mechanisms by which IRE1 regulates the UPR dependent on the stress stimulus. Moreover, IRE1 forms hetero-oligomers with other proteins, activating the JNK and ERK pathways, and directly interacts with ribosome complexes with potential implications in translational regulation and RNA decay, as well as with MAMs to dictate cellular energetics.
Clinically, regulation of the IRE1–XBP1 axis has been demonstrated in a model- and disease-dependent manner (Figure 2). While end-stage heart failure resulting from ischemic heart disease and dilated cardiomyopathy is associated with a sustained upregulation of UPR and the IRE1 pathway in patients, animal models reveal dynamic regulation of IRE1 signaling during progression to heart failure. HFpEF patients and a unique HFpEF mouse model displayed a suppression of UPR, and more specifically XBP1 splicing. Sustained XBP1 splicing is crucial to chronic progression of atherosclerosis by inducing endothelial dysfunction, vascular smooth muscle cell proliferation and migration, and macrophage-mediated inflammation. Conversely, sufficient and brief upregulation of XBP1 splicing in cardiomyocytes is essential for induction of UPR in the setting of ischemia/reperfusion and angiogenesis. In diabetic cardiomyopathy, pro-inflammatory pathways, such as the NLRP3 inflammasome, play a central role in disease progression. IRE1 has been shown to modulate the NLRP3 inflammasome in diabetic mice, although evidence of possible disease modulation by cardiac IRE1 is missing.
Figure 2.

Summary of mechanisms of the IRE1/XBP1 pathway in cardiovascular disease. (i) In atherosclerosis, GRP78, GRP94, and CHOP are upregulated in human samples. Increased spliced XBP1 (XBP1s) is present in dysfunctional endothelial cells, proliferating, migrating, and calcified vSMC. Unspliced XBP1 (XBP1u) is reduced in calcified vSMC. (ii) In cardiac ischemia/reperfusion, XBP1s is protective through induction of the hexosamine biosynthetic pathway, enhancing protein folding and preserving cardiac function. Additionally, XBP1s drives post-ischemic angiogenesis. (iii) In HFrEF patients, GRP78, GRP94, and calreticulin are upregulated. XBP1s induces BNP expression and drives fibrosis. In HFpEF patients, isolated downregulation of XBP1s is observed through inhibitory S-nitrosylation of IRE1. (iv) In diabetic cardiomyopathy, chronic inflammation is perpetuated by the NLRP3 inflammasome. The IRE1 pathway, which has been shown to induce NLRP3 in the pancreas, may play a role in NLRP3 regulation and chronic inflammation in diabetic cardiomyopathy.
Genetic approaches to modify the IRE1/XBP1 pathway in humans remain challenging due to the complexity of the IRE1/XBP1 signaling cascade and the transient nature of stress signaling, requiring timely control of activation and deactivation. Recent advances in deploying small molecules specifically targeting independent functions of IRE1 have shown positive effects in animal disease models. STF-083010 and 4μ8c are RNase inhibitors often used in tumor research, where inhibition of an overactive IRE1 branch blocks tumor progression. While IRE1 kinase inhibitors (type I) such as APY29, which stabilize an active kinase-domain conformation, activate the IRE1 RNase domain, other IRE1 kinase inhibitors (type II) such as KIRA3/6/7, which stabilize an inactive ATP-binding site conformation in IRE1, keep the RNase domain in an inactive state [97]. Since kinase-driven oligomerization of IRE1 hyperactivates its RNase to trigger apoptosis, then kinase inhibitors that block oligomerization (type II) should prevent apoptosis under ER stress. More intriguingly, recent work by the Wiseman lab identified a group of IRE1-specific activators, among which IXA4 and IXA6 showed the most potent induction in XBP1 splicing while maintaining little-to-no cytotoxicity [98]. IXA4 was successfully deployed in a diet-induced obese mouse model, where a brief boosting of IRE1/XBP1 signaling led to multitissue benefits without triggering the maladaptive UPR phase, providing a potential clinically relevant tool for the cardiovascular field [99].
The IRE1 pathway does not exist independently from the ATF6 and PERK pathways, as extensive crosstalk occurs between all three (Figure 1a). In the future, complex, tissue-specific high-throughput analysis approaches, up to single-cell resolution, will enable thorough characterization of the cardiac UPRosome, including the roles of the UPR in cellular crosstalk (Figure 2).
Funding
This work was supported by the German Centre for Cardiovascular Research (DZHK) Excellence Program, The University of Arizona Health Sciences Career Development Award Program, and the Translational Cardiovascular Research Center at the University of Arizona College of Medicine – Phoenix.
Footnotes
Conflict of interest statement
Nothing declared.
CRediT authorship contribution statement
F.F. and S.D. Conceptualization; S.D. Funding acquisition; S.D. Project administration; S.D. Supervision; F.F. Visualization; F.F. Writing – original draft; F.F. and S.D. Writing – review & editing.
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as
• of special interest
•• of outstanding interest
- 1.Lashkevich KA, Dmitriev SE: mRNA targeting, transport and local translation in eukaryotic cells: from the classical view to a diversity of new concepts. Mol Biol 2021, 55:507–537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Wong WL, Brostrom MA, Kuznetsov G, Gmitter-Yellen D, Brostrom CO: Inhibition of protein synthesis and early protein processing by thapsigargin in cultured cells. Biochem J 1993, 289:71–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Harding HP, Zhang Y, Ron D: Protein translation and folding are coupled by an endoplasmic- reticulum-resident kinase. Nature 1999, 397:271–274. [DOI] [PubMed] [Google Scholar]
- 4.DuRose JB, Tam AB, Niwa M: Intrinsic capacities of molecular sensors of the unfolded protein response to sense alternate forms of endoplasmic reticulum stress. Mol Biol Cell 2006, 17:3095–3107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Haze K, Yoshida H, Yanagi H, Yura T, Mori K: Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell 1999, 10:3787–3799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lin JH, Li H, Yasumura D, Cohen HR, Zhang C, Panning B, Shokat KM, Lavail MM, Walter P: IRE1 signaling affects cell fate during the unfolded protein response. Science 2007, 318:944–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Höhn A, Tramutola A, Cascella R: Proteostasis failure in neurodegenerative diseases: focus on oxidative stress. Oxid Med Cell Longev 2020, 2020:5497046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ottens F, Franz A, Hoppe T: Build-UPS and break-downs: metabolism impacts on proteostasis and aging. Cell Death Differ 2021, 28:505–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hofmann C, Katus HA, Doroudgar S: Protein misfolding in cardiac disease. Circulation 2019, 139:2085–2088. [DOI] [PubMed] [Google Scholar]
- 10.Hetz C, Papa FR: The unfolded protein response and cell fate control. Mol Cell 2018, 69:169–181. [DOI] [PubMed] [Google Scholar]
- 11.Elia AE, Lalli S, Monsurrò MR, Sagnelli A, Taiello AC, Reggiori B, La Bella V, Tedeschi G, Albanese A: Tauroursodeoxycholic acid in the treatment of patients with amyotrophic lateral sclerosis. Eur J Neurol 2016, 23:45–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Arkwright R, Pham TM, Zonder JA, Dou QP: The preclinical discovery and development of bortezomib for the treatment of mantle cell lymphoma. Expert Opin Drug Discov 2017, 12:225–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hetz C, Axten JM, Patterson JB: Pharmacological targeting of the unfolded protein response for disease intervention. Nat Chem Biol 2019, 15:764–775. [DOI] [PubMed] [Google Scholar]
- 14.Hetz C, Chevet E, Oakes SA: Proteostasis control by the unfolded protein response. Nat Cell Biol 2015, 17:829–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.van Ziel AM, Scheper W: The UPR in neurodegenerative disease: not just an inside job. Biomolecules (8) 2020, 10:1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cox JS, Shamu CE, Walter P: Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase. Cell 1993, 73:1197–1206. [DOI] [PubMed] [Google Scholar]
- 17.Morl K, Ma W, Gething M-JJ, Sambrook J, Mori K, Ma W, Gething M-JJ, Sambrook J: A transmembrane protein with a cdc2+ CDC28-related kinase activity is required for signaling from the ER to the nucleus. Cell 1993, 74:743–756. [DOI] [PubMed] [Google Scholar]
- 18.Hollien J: Evolution of the unfolded protein response. Biochim Biophys Acta Mol Cell Res 2013, 1833:2458–2463. [DOI] [PubMed] [Google Scholar]
- 19.Bertolotti A, Wang XZ, Novoa I, Jungreis R, Schlessinger K, Cho JH, West AB, Ron D: Increased sensitivity to dextran sodium sulfate colitis in IRE1β-deficient mice. J Clin Investig 2001, 107:585–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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, 2:326–332. [DOI] [PubMed] [Google Scholar]
- 21.Carrara M, Prischi F, Nowak PR, Kopp MC, Ali MMU: Noncanonical binding of BiP ATPase domain to Ire1 and Perk is dissociated by unfolded protein CH1 to initiate ER stress signaling. Elife 2015, 4:e03522.(2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kimata Y, Oikawa D, Shimizu Y, Ishiwata-Kimata Y, Kohno K: A role for BiP as an adjustor for the endoplasmic reticulum stress-sensing protein Ire1. J Cell Biol 2004, 167:445–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Credle JJ, Finer-Moore JS, Papa FR, Stroud RM, Walter P: On the mechanism of sensing unfolded protein in the endoplasmic reticulum. Proc Natl Acad Sci U S A 2005, 102:18773–18784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sundaram A, Appathurai S, Plumb R, Mariappan M: Dynamic changes in complexes of IRE1α, PERK, and ATF6α during endoplasmic reticulum stress. Mol Biol Cell 2018, 29:1376–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Karagöz GE, Acosta-Alvear D, Nguyen HT, Lee CP, Chu F, Walter P: An unfolded protein-induced conformational switch activates mammalian IRE1. Elife 2017, 6:e30700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gardner BM, Walter P: Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response. Science 2011, 333:1891–1894 (80-). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hetz C, Glimcher LH: Fine tuning of the unfolded protein response: assembling the IRE1α interactome. Mol Cell 2009, 35:551–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Karagöz GE, Aragón T, Acosta-Alvear D: Recent advances in signal integration mechanisms in the unfolded protein response. F1000Research 2019, 8:F1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Korennykh AV, Egea PF, Korostelev AA, Finer-Moore J, Zhang C, Shokat KM, Stroud RM, Walter P: The unfolded protein response signals through high-order assembly of Ire1. Nature 2009, 457:687–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lee KPK, Dey M, Neculai D, Cao C, Dever TE, Sicheri F: Structure of the dual enzyme Ire1 reveals the basis for catalysis and regulation in non-conventional RNA splicing. Cell 2008, 132:89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K: XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001, 107:881–891. [DOI] [PubMed] [Google Scholar]
- 32.Lee A-H, Iwakoshi NN, Glimcher LH: XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 2003, 23:7448–7459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yanagitani K, Imagawa Y, Iwawaki T, Hosoda A, Saito M, Kimata Y, Kohno K: Cotranslational targeting of XBP1 protein to the membrane promotes cytoplasmic splicing of its own mRNA. Mol Cell 2009, 34:191–200. [DOI] [PubMed] [Google Scholar]
- 34.Hollien J, Weissman JS: Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science 2006, 313:104–107 (80-). [DOI] [PubMed] [Google Scholar]
- 35.Lu M, Lawrence DA, Marsters S, Acosta-Alvear D, Kimmig P, Mendez AS, Paton AW, Paton JC, Walter P, Ashkenazi A: Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science 2014, 345:98–101 (80-). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Han D, Lerner AG, Vande Walle L, Upton JP, Xu W, Hagen A, Backes BJ, Oakes SA, Papa FR: IRE1α kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell 2009, 138:562–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37 ••.Le Thomas A, Ferri E, Marsters S, Harnoss JM, Lawrence DA, Zuazo-Gaztelu I, Modrusan Z, Chan S, Solon M, Chalouni C, et al. : Decoding non-canonical mRNA decay by the endoplasmic-reticulum stress sensor IRE1$α$. Nat Commun (1) 2021, 12:7310. [DOI] [PMC free article] [PubMed] [Google Scholar]; Novel non-canonical IRE1 interactions and functions.
- 38.Lhomond S, Avril T, Dejeans N, Voutetakis K, Doultsinos D, McMahon M, Pineau R, Obacz J, Papadodima O, Jouan F, et al. : Dual IRE1 RNase functions dictate glioblastoma development. EMBO Mol Med 2018, 10:e7929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nishitoh H, Matsuzawa A, Tobiume K, Saegusa K, Takeda K, Inoue K, Hori S, Kakizuka A, Ichijo H: ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev 2002, 16:1345–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Urano F, Wang XZ, Bertolotti A, Zhang Y, Chung P, Harding HP, Ron D: Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000, 287:664–666 (80-). [DOI] [PubMed] [Google Scholar]
- 41.Nguyên DT, Kebache S, Fazel A, Wong HN, Jenna S, Emadali A, Lee EH, Bergeron JJM, Kaufman RJ, Larose L, et al. : Nck-dependent activation of extracellular signal-regulated kinase-1 and regulation of cell survival during endoplasmic reticulum stress. Mol Biol Cell 2004, 15:4248–4260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yoshida H, Oku M, Suzuki M, Mori K: pXBP1(U) encoded in XBP1 pre-mRNA negatively regulates unfolded protein response activator pXBP1(S) in mammalian ER stress response. J Cell Biol 2006, 172:565–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yoshida H, Uemura A, Mori K: pXBP1(U), a negative regulator of the unfolded protein response activator pXBP1(S), targets ATF6 but not ATF4 in proteasome-mediated degradation. Cell Struct Funct 2009, 34:1–10. [DOI] [PubMed] [Google Scholar]
- 44.Zhao Y, Li X, Cai MY, Ma K, Yang J, Zhou J, Fu W, Wei FZ, Wang L, Xie D, et al. : XBP-1u suppresses autophagy by promoting the degradation of FoxO1 in cancer cells. Cell Res 2013, 23:491–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Martin D, Li Y, Yang J, Wang G, Margariti A, Jiang Z, Yu H, Zampetaki A, Hu Y, Xu Q, et al. : Unspliced X-box-binding protein 1 (XBP1) protects endothelial cells from oxidative stress through interaction with histone deacetylase 3. J Biol Chem 2014, 289:30625–30634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Acosta-Alvear D, Karagöz GE, Fröhlich F, Li H, Walther TC, Walter P: The unfolded protein response and endoplasmic reticulum protein targeting machineries converge on the stress sensor IRE1. Elife 2018, 7:e43036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47 •.Carreras-Sureda A, Jaña F, Urra H, Durand S, Mortenson DE, Sagredo A, Bustos G, Hazari Y, Ramos-Fernández E, Sassano ML, et al. : Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat Cell Biol 2019, 21:755–767. [DOI] [PMC free article] [PubMed] [Google Scholar]; Novel non-canonical IRE1 interactions and functions.
- 48.Taylor RC, Dillin A: XBP-1 is a cell-nonautonomous regulator of stress resistance and longevity. Cell (7) 2013, 153:1435–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Eschenhagen T, Bolli R, Braun T, Field LJ, Fleischmann BK, Frisén J, Giacca M, Hare JM, Houser S, Lee RT, et al. : Cardiomyocyte regeneration: a consensus statement. Circulation 2017, 136:680–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ren J, Bi Y, Sowers JR, Hetz C, Zhang Y: Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases. Nat Rev Cardiol 2021, 18:499–521. [DOI] [PubMed] [Google Scholar]
- 51.Abbafati C, Abbas KM, Abbasi-Kangevari M, Abd-Allah F, Abdelalim A, Abdollahi M, Abdollahpour I, Abegaz KH, Abolhassani H, Aboyans V, et al. : Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396:1204–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Doroudgar S, Thuerauf DJDJ, Marcinko MCMC, Belmont PJPJ, Glembotski CCC: Ischemia activates the ATF6 branch of the endoplasmic reticulum stress response*. J Biol Chem 2009, 284:29735–29745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Glembotski CC, Rosarda JD, Wiseman RL: Proteostasis and beyond: ATF6 in ischemic disease. Trends Mol Med 2019, 25:538–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Glembotski CC, Arrieta A, Blackwood EA, Stauffer WT: ATF6 as a nodal regulator of proteostasis in the heart. Front Physiol 2020, 11:267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang S, Binder P, Fang Q, Wang Z, Xiao W, Liu W, Wang X: Endoplasmic reticulum stress in the heart: insights into mechanisms and drug targets. Br J Pharmacol 2018, 175:1293–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, Burri H, Butler J, Celutkiene J, Chioncel O, et al. : 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J 2021, 42:3599–3726. [DOI] [PubMed] [Google Scholar]
- 57.Reichardt IM, Robeson KZ, Regnier M, Davis J: Controlling cardiac fibrosis through fibroblast state space modulation. Cell Signal 2021, 79:109888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Liu T, Song D, Dong J, Zhu P, Liu J, Liu W, Ma X, Zhao L, Ling S: Current understanding of the pathophysiology of myocardial fibrosis and its quantitative assessment in heart failure. Front Physiol 2017, 8:238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Park S-M, Kang T-I, So J-S: Roles of XBP1s in transcriptional regulation of target genes. Biomed 2021, 9:791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Steiger DA, Yokota T, Li J, Ren S, Minamisawa S, Wang Y: The serine/threonine-protein kinase/endoribonuclease IRE1α protects the heart against pressure overload–induced heart failure. J Biol Chem 2018, 293:9652–9661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Groenendyk J, Lee D, Jung J, Dyck JRB, Lopaschuk GD, Agellon LB, Michalak M: Inhibition of the unfolded protein response mechanism prevents cardiac fibrosis. PLoS One 2016, 11:e0159682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Qu J, Li M, Li D, Xin Y, Li J, Lei S, Wu W, Liu X: Stimulation of Sigma-1 receptor protects against cardiac fibrosis by alleviating IRE1 pathway and autophagy impairment. Oxid Med Cell Longev 2021, 2021:8836818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ortega A, Roselló-Lletí E, Tarazón E, Molina-Navarro MM, Martínez-Dolz L, González-Juanatey JR, Lago F, Montoro-Mateos JD, Salvador A, Rivera M, et al. : Endoplasmic reticulum stress induces different molecular structural alterations in human dilated and ischemic cardiomyopathy. PLoS One 2014, 9:e107635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Okada KI, Minamino T, Tsukamoto Y, Liao Y, Tsukamoto O, Takashima S, Hirata A, Fujita M, Nagamachi Y, Nakatani T, et al. : Prolonged endoplasmic reticulum stress in hypertrophic and failing heart after aortic constriction: possible contribution of endoplasmic reticulum stress to cardiac myocyte apoptosis. Circulation 2004, 110:705–712. [DOI] [PubMed] [Google Scholar]
- 65.Castillero E, Akashi H, Pendrak K, Yerebakan H, Najjar M, Wang C, Naka Y, Mancini D, Sweeney HL, D’Armiento J, et al. : Attenuation of the unfolded protein response and endoplasmic reticulum stress after mechanical unloading in dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2015, 309:H459–H470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Duan Q, Yang L, Gong W, Chaugai S, Wang F, Chen C, Wang P, Zou MH, Wang DW: MicroRNA-214 is upregulated in heart failure patients and suppresses XBP1-mediated endothelial cells angiogenesis. J Cell Physiol 2015, 230:1964–1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Duan Q, Chen C, Yang L, Li N, Gong W, Li S, Wang W: MicroRNA regulation of unfolded protein response transcription factor XBP1 in the progression of cardiac hypertrophy and heart failure in vivo. J Transl Med 2015, 13:363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Sawada T, Minamino T, Fu HY, Asai M, Okuda K, Isomura T, Yamazaki S, Asano Y, Okada K ichiro, Tsukamoto O, et al. : X-box binding protein 1 regulates brain natriuretic peptide through a novel AP1/CRE-like element in cardiomyocytes. J Mol Cell Cardiol 2010, 48:1280–1289. [DOI] [PubMed] [Google Scholar]
- 69 •.Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, Luo X, Jiang N, May HI, Wang ZV, et al. : Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019, 568:351–356. [DOI] [PMC free article] [PubMed] [Google Scholar]; Direct and strong evidence of involvement of IRE1-XBP1 in HFpEF.
- 70 ••.Hahn VS, Knutsdottir H, Luo X, Bedi K, Margulies KB, Haldar SM, Stolina M, Yin J, Khakoo AY, Vaishnav J, et al. : Myocardial gene expression signatures in human heart failure with preserved ejection fraction. Circulation 2021, 143:120–134. [DOI] [PMC free article] [PubMed] [Google Scholar]; Direct and strong evidence of involvement of IRE1-XBP1 in HFpEF.
- 71.Soehnlein O, Libby P: Targeting inflammation in atherosclerosis — from experimental insights to the clinic. Nat Rev Drug Discov 2021, 20:589–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Falk E: Pathogenesis of atherosclerosis. J Am Coll Cardiol 2006, 47:436–441. [DOI] [PubMed] [Google Scholar]
- 73.Libby P: Mechanisms of acute coronary syndromes and their implications for therapy. N Engl J Med 2013, 368:2004–2013. [DOI] [PubMed] [Google Scholar]
- 74.Myoishi M, Hao H, Minamino T, Watanabe K, Nishihira K, Hatakeyama K, Asada Y, Okada KI, Ishibashi-Ueda H, Gabbiani G, et al. : Increased endoplasmic reticulum stress in atherosclerotic plaques associated with acute coronary syndrome. Circulation 2007, 116:1226–1233. [DOI] [PubMed] [Google Scholar]
- 75.Zeng L, Zampetaki A, Margariti A, Pepe AE, Alam S, Martin D, Xiao Q, Wang W, Jin ZG, Cockerill G, et al. : Sustained activation of XBP1 splicing leads to endothelial apoptosis and atherosclerosis development in response to disturbed flow. Proc Natl Acad Sci U S A 2009, 106:8326–8331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zeng L, Li Y, Yang J, Wang G, Margariti A, Xiao Q, Zampetaki A, Yin X, Mayr M, Mori K, et al. : XBP 1-deficiency abrogates neointimal lesion of injured vessels via cross talk with the PDGF signaling. Arterioscler Thromb Vasc Biol 2015, 35:2134–2144. [DOI] [PubMed] [Google Scholar]
- 77.Liberman M, Johnson RC, Handy DE, Loscalzo J, Leopold JA: Bone morphogenetic protein-2 activates NADPH oxidase to increase endoplasmic reticulum stress and human coronary artery smooth muscle cell calcification. Biochem Biophys Res Commun 2011, 413:436–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yang L, Dai R, Wu H, Cai Z, Xie N, Zhang X, Shen Y, Gong Z, Jia Y, Yu F, et al. : Unspliced XBP1 counteracts β-catenin to inhibit vascular calcification. Circ Res 2022, 130:213–229. [DOI] [PubMed] [Google Scholar]
- 79.Tufanli O, Telkoparan Akillilar P, Acosta-Alvear D, Kocaturk B, Onat UI, Hamid SM, Çimen I, Walter P, Weber C, Erbay E, et al. : Targeting IRE1 with small molecules counteracts progression of atherosclerosis. Proc Natl Acad Sci 2017, 114:E1395–E1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Thuerauf DJ, Marcinko M, Gude N, Rubio M, Sussman MA, Glembotski CC: Activation of the unfolded protein response in infarcted mouse heart and hypoxic cultured cardiac myocytes. Circ Res 2006, 99:275–282, 10.1161/01.RES.0000233317.70421.03 [DOI] [PubMed] [Google Scholar]
- 81.Wang ZV, Deng Y, Gao N, Pedrozo Z, Li DL, Morales CR, Criollo A, Luo X, Tan W, Jiang N, et al. : Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell 2014, 156:1179–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zeng L, Xiao Q, Chen M, Margariti A, Martin D, Ivetic A, Xu H, Mason J, Wang W, Cockerill G, et al. : Vascular endothelial cell growth-activated XBP1 splicing in endothelial cells is crucial for angiogenesis. Circulation 2013, 127:1712–1722. [DOI] [PubMed] [Google Scholar]
- 83.Borghetti G, Von Lewinski D, Eaton DM, Sourij H, Houser SR, Wallner M: Diabetic cardiomyopathy: current and future therapies. Beyond glycemic control. Front Physiol 2018, 9:1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lopaschuk GD, Verma S: Mechanisms of cardiovascular benefits of sodium glucose co-transporter 2 (SGLT2) inhibitors: a state-of-the-art review. Basic to Transl Sci 2020, 5:632–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tan Y, Zhang Z, Zheng C, Wintergerst KA, Keller BB, Cai L: Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol 2020, 17:585–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lemmer IL, Willemsen N, Hilal N, Bartelt A: A guide to understanding endoplasmic reticulum stress in metabolic disorders. Mol Metab 2021, 47:101169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wu R, Zhang QH, Lu YJ, Ren K, Yi GH: Involvement of the IRE1α-XBP1 pathway and XBP1s-dependent transcriptional reprogramming in metabolic diseases. DNA Cell Biol 2015, 34:6–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88 ••.Madhavan A, Kok BP, Rius B, Grandjean JMD, Alabi A, Albert V, Sukiasyan A, Powers ET, Galmozzi A, Saez E, et al. : Pharmacologic IRE1/XBP1s activation promotes systemic adaptive remodeling in obesity. Nat Commun 2022, 13:1–9 (2022 131). [DOI] [PMC free article] [PubMed] [Google Scholar]; Pharmacological activation of the IRE1-XBP1 axis achieves multiorgan benefits in obesity.
- 89.Luo B, Li B, Wang W, Liu X, Xia Y, Zhang C, Zhang M, Zhang Y, An F: NLRP3 gene silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model. PLoS One 2014, 9:e104771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Luo B, Li B, Wang W, Liu X, Liu X, Xia Y, Zhang C, Zhang Y, Zhang M, An F: Rosuvastatin alleviates diabetic cardiomyopathy by inhibiting NLRP3 inflammasome and MAPK pathways in a type 2 diabetes rat model. Cardiovasc Drugs Ther 2014, 28:33–43. [DOI] [PubMed] [Google Scholar]
- 91.Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y: SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with type 2 diabetes. Further augmentation of the effects with saxagliptin, a DPP4 inhibitor. Cardiovasc Drugs Ther 2017, 31:119–132. [DOI] [PubMed] [Google Scholar]
- 92.Chen H, Tran D, Yang HC, Nylander S, Birnbaum Y, Ye Y: Dapagliflozin and ticagrelor have additive effects on the attenuation of the activation of the NLRP3 inflammasome and the progression of diabetic cardiomyopathy: an AMPK–mTOR interplay. Cardiovasc Drugs Ther 2020, 34:443–461. [DOI] [PubMed] [Google Scholar]
- 93.Elmadbouh I, Singla DK: Bmp-7 attenuates inflammation-induced pyroptosis and improves cardiac repair in diabetic cardiomyopathy. Cells (10) 2021, 10:2640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lerner AG, Upton JP, Praveen PVK, Ghosh R, Nakagawa Y, Igbaria A, Shen S, Nguyen V, Backes BJ, Heiman M, et al. : IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab 2012, 16:250–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Mori K: The unfolded protein response: the dawn of a new field. Proc Jpn Acad Ser B Phys Biol Sci 2015, 91:469–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Albert and Mary Lasker Foundation: Unfolded protein response - 2014 Albert Lasker Basic Medical Research Award. 〈https://laskerfoundation.org/winners/unfolded-protein-response/〉 2014,
- 97.Langlais T, Pelizzari-Raymundo D, Mahdizadeh SJ, Gouault N, Carreaux F, Chevet E, Eriksson LA, Guillory X: Structural and molecular bases to IRE1 activity modulation. Biochem J 2021, 478:2953–2975. [DOI] [PubMed] [Google Scholar]
- 98.Grandjean JMD, Madhavan A, Cech L, Seguinot BO, Paxman RJ, Smith E, Scampavia L, Powers ET, Cooley CB, Plate L, et al. : Pharmacologic IRE1/XBP1s activation confers targeted ER proteostasis reprogramming. Nat Chem Biol 2020, 16:1052–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Madhavan A, Kok BP, Rius B, Grandjean JMD, Alabi A, Albert V, Sukiasyan A, Powers ET, Galmozzi A, Saez E, et al. : Pharmacologic IRE1/XBP1s activation promotes systemic adaptive remodeling in obesity. Nat Commun 2022, 13:608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Yan W, Frank CL, Korth MJ, Sopher BL, Novoa I, Ron D, Katze MG: Control of PERK elF2α kinase activity by the endoplasmic reticulum stress-induced molecular chaperone P58IPK. Proc Natl Acad Sci U S A 2002, 99:15920–15925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chang TK, Lawrence DA, Lu M, Tan J, Harnoss JM, Marsters SA, Liu P, Sandoval W, Martin SE, Ashkenazi A: Coordination between two branches of the unfolded protein response determines apoptotic cell fate. Mol Cell 2018, 71:629–636 e5. [DOI] [PubMed] [Google Scholar]
- 102.Lee J, Sun C, Zhou Y, Lee J, Gokalp D, Herrema H, Park SW, Davis RJ, Ozcan U: P38 MAPK-mediated regulation of Xbp1s is crucial for glucose homeostasis. Nat Med 2011, 17:1251–1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Park SW, Zhou Y, Lee J, Lu A, Sun C, Chung J, Ueki K, Ozcan U: The regulatory subunits of PI3K, p85α and p85β, interact with XBP-1 and increase its nuclear translocation. Nat Med 2010. 164 2010, 16:429–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yamamoto K, Yoshida H, Kokame K, Kaufman RJ, Mori K: Differential contributions of ATF6 and XBP1 to the activation of endoplasmic reticulum stress-responsive cis-acting elements ERSE, UPRE and ERSE-II. J Biochem 2004, 136:343–350. [DOI] [PubMed] [Google Scholar]
- 105.Yoneda T, Imaizumi K, Oono K, Yui D, Gomi F, Katayama T, Tohyama M: Activation of caspase-12, an endoplastic reticulum (ER) resident caspase, through tumor necrosis factor receptor-associated factor 2-dependent mechanism in response to the ER stress. J Biol Chem 2001, 276:13935–13940. [DOI] [PubMed] [Google Scholar]
- 106.Marcu MG, Doyle M, Bertolotti A, Ron D, Hendershot L, Neckers L: Heat shock protein 90 modulates the unfolded protein response by stabilizing IRE1alpha. Mol Cell Biol 2002, 22:8506–8513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Gu F, Duc TN, Stuible M, Dubé N, Tremblay ML, Chevet E: Protein-tyrosine phosphatase 1B potentiates IRE1 signaling during endoplasmic reticulum stress. J Biol Chem 2004, 279:49689–49693. [DOI] [PubMed] [Google Scholar]
- 108.Yang Q, Kim YS, Lin Y, Lewis J, Neckers L, Liu ZG: Tumour necrosis factor receptor 1 mediates endoplasmic reticulum stress-induced activation of the MAP kinase JNK. EMBO Rep 2006, 7:622–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hetz C, Bernasconi P, Fisher J, Lee AH, Bassik MC, Antonsson B, Brandt GS, Iwakoshi NN, Schrinzel A, Glimcher LH, et al. : Proapoptotic BAX and BAK modulate the unfolded protein response by a direct interaction with IRE1α. Science 2006, 312:572–576 (80-). [DOI] [PubMed] [Google Scholar]
- 110.Hu P, Han Z, Couvillon AD, Kaufman RJ, Exton JH: Autocrine tumor necrosis factor alpha links endoplasmic reticulum stress to the membrane death receptor pathway through IRE1alpha-mediated NF-kappaB activation and downregulation of TRAF2 expression. Mol Cell Biol 2006, 26:3071–3084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Luo D, He Y, Zhang H, Yu L, Chen H, Xu Z, Tang S, Urano F, Min W: AIP1 is critical in transducing IRE1-mediated endoplasmic reticulum stress response. J Biol Chem 2008, 283:11905–11912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gupta S, Deepti A, Deegan S, Lisbona F, Hetz C, Samali A: HSP72 protects cells from ER stress-induced apoptosis via enhancement of IRE1α-XBP1 signaling through a physical interaction. PLoS Biol 2010, 8:1000410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Rodriguez DA, Zamorano S, Lisbona F, Rojas-Rivera D, Urra H, Cubillos-Ruiz JR, Armisen R, Henriquez DR, Cheng EH, Letek M, et al. : BH3-only proteins are part of a regulatory network that control the sustained signalling of the unfolded protein response sensor IRE1α. EMBO J 2012, 31:2322–2335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.He Y, Beatty A, Han X, Ji Y, Ma X, Adelstein RS, Yates JR, Kemphues K, Qi L: Nonmuscle Myosin IIB links cytoskeleton to IRE1α signaling during ER stress. Dev Cell 2012, 23:1141–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Jwa M, Chang P: PARP16 is a tail-anchored endoplasmic reticulum protein required for the PERK-and IRE1α-mediated unfolded protein response. Nat Cell Biol 2012, 14:1223–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Arshad M, Ye Z, Gu X, Wong CK, Liu Y, Li D, Zhou L, Zhang Y, Bay WP, Yu VC, et al. : RNF13, a RING finger protein, mediates endoplasmic reticulum stress-induced apoptosis through the inositol-requiring enzyme (IRE1α)/c-Jun NH2-terminal kinase pathway. J Biol Chem 2013, 288:8726–8736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mori T, Hayashi T, Hayashi E, Su TP: Sigma-1 receptor chaperone at the ER-mitochondrion interface mediates the mitochondrion-ER-nucleus signaling for cellular survival. PLoS One 2013, 8:e76941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Chen L, Xu S, Liu L, Wen X, Xu Y, Chen J, Teng J: Cab45S inhibits the ER stress-induced IRE1-JNK pathway and apoptosis via GRP78/BiP. Cell Death Dis 2014, 5:e1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhu X, Zhang J, Sun H, Jiang C, Dong Y, Shan Q, Su S, Xie Y, Xu N, Lou X, et al. : Ubiquitination of inositol-requiring enzyme 1 (IRE1) by the E3 ligase CHIP mediates the IRE1/TRAF2/JNK pathway. J Biol Chem 2014, 289:30567–30577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Morita S, Villalta SA, Feldman HC, Register AC, Rosenthal W, Hoffmann-Petersen IT, Mehdizadeh M, Ghosh R, Wang L, Colon-Negron K, et al. : Targeting ABL-IRE1α signaling spares ER-stressed pancreatic β cells to reverse autoimmune diabetes. Cell Metab 2017, 25:883–897 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Tavernier Q, Bennana E, Poindessous V, Schaeffer C, Rampoldi L, Pietrancosta N, Pallet N: Regulation of IRE1 RNase activity by the Ribonuclease inhibitor 1 (RNH1). Cell Cycle 2018, 17:1901–1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sepulveda D, Rojas-Rivera D, Rodríguez DA, Groenendyk J, Köhler A, Lebeaupin C, Ito S, Urra H, Carreras-Sureda A, Hazari Y, et al. : Interactome screening identifies the ER luminal chaperone Hsp47 as a regulator of the unfolded protein response transducer IRE1α. Mol Cell 2018, 69:238–252 e7. [DOI] [PubMed] [Google Scholar]
- 123.Wu IH, Yoon JS, Yang Q, Liu Y, Skach W, Thomas P: A role for the ribosome-associated complex in activation of the IRE1 branch of UPR. Cell Rep 2021, 35:109217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Shin GC, Moon SU, Kang HS, Choi HS, Han HD, Kim KH: PRKCSH contributes to tumorigenesis by selective boosting of IRE1 signaling pathway. Nat Commun 2019, 10:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Welihinda AA, Tirasophon W, Green SR, Kaufman RJ: Protein serine/threonine phosphatase Ptc2p negatively regulates the unfolded-protein response by dephosphorylating Ire1p kinase. Mol Cell Biol 1998, 18:1967–1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Oono K, Yoneda T, Manabe T, Yamagishi S, Matsuda S, Hitomi J, Miyata S, Mizuno T, Imaizumi K, Katayama T, et al. : JAB1 participates in unfolded protein responses by association and dissociation with IRE1. Neurochem Int 2004, 45:765–772. [DOI] [PubMed] [Google Scholar]
- 127.Guo J, Polymenis M: Dcr2 targets Ire1 and downregulates the unfolded protein response in Saccharomyces cerevisiae. EMBO Rep 2006, 7:1124–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gao B, Lee SM, Chen A, Zhang J, Zhang DD, Kannan K, Ortmann RA, Fang D: Synoviolin promotes IRE1 ubiquitination and degradation in synovial fibroblasts from mice with collagen-induced arthritis. EMBO Rep 2008, 9:480–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lisbona F, Rojas-Rivera D, Thielen P, Zamorano S, Todd D, Martinon F, Glavic A, Kress C, Lin JH, Walter P, et al. : BAX inhibitor-1 Is a negative regulator of the ER stress sensor IRE1a fernanda. Mol Cell 2009, 33:679–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Nagai A, Kadowaki H, Maruyama T, Takeda K, Nishitoh H, Ichijo H: USP14 inhibits ER-associated degradation via interaction with IRE1α. Biochem Biophys Res Commun 2009, 379:995–1000. [DOI] [PubMed] [Google Scholar]
- 131.Qiu Y, Mao T, Zhang Y, Shao M, You J, Ding Q, Chen Y, Wu D, Xie D, Lin X, et al. : A crucial role for RACK1 in the regulation of glucose-stimulated IRE1α activation in pancreatic β cells. Sci Signal (106) 2010, 3:ra7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lu G, Ota A, Ren S, Franklin S, Rau CD, Ping P, Lane TF, Zhou ZH, Reue K, Lusis AJ, et al. : PPM1l encodes an inositol requiring-protein 1 (IRE1) specific phosphatase that regulates the functional outcome of the ER stress response. Mol Metab 2013, 2:405–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ren S, Lu G, Ota A, Zhou ZH, Vondriska TM, Lane TF, Wang Y: IRE1 phosphatase PP2Ce regulates adaptive ER stress response in the postpartum mammary gland. PLoS One (11) 2014, 9:e111606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Eletto D, Eletto D, Dersh D, Gidalevitz T, Argon Y: Protein disulfide isomerase A6 controls the decay of IRE1α signaling via disulfide-dependent association. Mol Cell 2014, 53:562–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Brozzi F, Gerlo S, Grieco FA, Nardelli TR, Lievens S, Gysemans C, Marselli L, Marchetti P, Mathieu C, Tavernier J, et al. : A combined “omics” approach identifies N-Myc interactor as a novel cytokine-induced regulator of IRE1α protein and c-Jun N-terminal kinase in pancreatic beta cells. J Biol Chem 2014, 289:20677–20693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Brozzi F, Gerlo S, Grieco FA, Juusola M, Balhuizen A, Lievens S, Gysemans C, Bugliani M, Mathieu C, Marchetti P, et al. : Ubiquitin D regulates IRE1α/c-Jun N-terminal Kinase (JNK) protein-dependent apoptosis in pancreatic beta cells. J Biol Chem 2016, 291:12040–12056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Sundaram A, Plumb R, Appathurai S, Mariappan M: The Sec61 translocon limits IRE1α signaling during the unfolded protein response. Elife 2017, 6:e27187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Plumb R, Zhang ZR, Appathurai S, Mariappan M: A functional link between the co-translational protein translocation pathway and the UPR. Elife 2015, 4:2–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Amin-Wetzel N, Saunders RA, Kamphuis MJ, Rato C, Preissler S, Harding HP, Ron D, J-Protein A: Co-chaperone recruits BiP to monomerize IRE1 and repress the unfolded protein response. Cell 2017, 171:1625–1637.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Pinkaew D, Chattopadhyay A, King MD, Chunhacha P, Liu Z, Stevenson HL, Chen Y, Sinthujaroen P, McDougal OM, Fujise K: Fortilin binds IRE1$α$ and prevents ER stress from signaling apoptotic cell death. Nat Commun 2017, 8:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wang Q, Groenendyk J, Paskevicius T, Qin W, Kor KC, Liu Y, Hiess F, Knollmann BC, Chen SRW, Tang J, et al. : Two pools of IRE1α in cardiac and skeletal muscle cells. FASEB J 2019, 33:8892–8904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Li X, Sun S, Appathurai S, Sundaram A, Plumb R, Mariappan M: A molecular mechanism for turning off IRE1α signaling during endoplasmic reticulum stress. Cell Rep 2020, 33:108563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Grey MJ, Cloots E, Simpson MS, LeDuc N, Serebrenik YV, De Luca H, De Sutter D, Luong P, Thiagarajah JR, Paton AW, et al. : IRE1β negatively regulates IRE1α signaling in response to endoplasmic reticulum stress. J Cell Biol 2020, 219:e201904048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Xu Y, Chen J, Chen J, Teng J: EI24 promotes cell adaption to ER stress by coordinating IRE1 signaling and calcium homeostasis. EMBO Rep 2022, 23:e51679, 10.15252/EMBR.202051679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Urra H, Henriquez DR, Cánovas J, Villarroel-Campos D, Carreras-Sureda A, Pulgar E, Molina E, Hazari YM, Limia CM, Alvarez-Rojas S, et al. : IRE1α governs cytoskeleton remodelling and cell migration through a direct interaction with filamin A. Nat Cell Biol 2018, 20:942–953. [DOI] [PubMed] [Google Scholar]
