Next Article in Journal
Surface Modification of Basalt Fibres with ZnO Nanorods and Its Effect on Thermal and Mechanical Properties of PLA-Based Composites
Next Article in Special Issue
Signaling Nodes Associated with Endoplasmic Reticulum Stress during NAFLD Progression
Previous Article in Journal
Different Aggregation Pathways and Structures for Aβ40 and Aβ42 Peptides
Previous Article in Special Issue
ER Stress-Sensor Proteins and ER-Mitochondrial Crosstalk—Signaling Beyond (ER) Stress Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

ER Stress and Unfolded Protein Response in Leukemia: Friend, Foe, or Both?

by
Kelly Féral
1,2,†,
Manon Jaud
1,2,†,
Céline Philippe
3,†,
Doriana Di Bella
3,
Stéphane Pyronnet
1,2,
Kevin Rouault-Pierre
3,
Laurent Mazzolini
1,4,*,‡ and
Christian Touriol
1,2,*,‡
1
Inserm UMR1037-Cancer Research Center of Toulouse, 2 avenue Hubert Curien, Oncopole entrée C, CS 53717, 31037 Toulouse, France
2
Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France
3
Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK
4
CNRS ERL5294, CRCT, F-31037 Toulouse, France
*
Authors to whom correspondence should be addressed.
These three authors contributed equally to this work.
These two authors share senior authorship.
Biomolecules 2021, 11(2), 199; https://doi.org/10.3390/biom11020199
Submission received: 15 December 2020 / Revised: 19 January 2021 / Accepted: 25 January 2021 / Published: 30 January 2021
(This article belongs to the Special Issue Endoplasmic Reticulum Stress in Diseases)

Abstract

:
The unfolded protein response (UPR) is an evolutionarily conserved adaptive signaling pathway triggered by a stress of the endoplasmic reticulum (ER) lumen compartment, which is initiated by the accumulation of unfolded proteins. This response, mediated by three sensors-Inositol Requiring Enzyme 1 (IRE1), Activating Transcription Factor 6 (ATF6), and Protein Kinase RNA-Like Endoplasmic Reticulum Kinase (PERK)—allows restoring protein homeostasis and maintaining cell survival. UPR represents a major cytoprotective signaling network for cancer cells, which frequently experience disturbed proteostasis owing to their rapid proliferation in an usually unfavorable microenvironment. Increased basal UPR also participates in the resistance of tumor cells against chemotherapy. UPR activation also occurs during hematopoiesis, and growing evidence supports the critical cytoprotective role played by ER stress in the emergence and proliferation of leukemic cells. In case of severe or prolonged stress, pro-survival UPR may however evolve into a cell death program called terminal UPR. Interestingly, a large number of studies have revealed that the induction of proapoptotic UPR can also strongly contribute to the sensitization of leukemic cells to chemotherapy. Here, we review the current knowledge on the consequences of the deregulation of UPR signaling in leukemias and their implications for the treatment of these diseases.

1. Introduction

About one-third of human genes encode secreted or transmembrane proteins as well as proteins resident of the endoplasmic reticulum, the Golgi apparatus, and lysosomes. Most of these proteins are targeted to the ER. The endoplasmic reticulum is a complex network of membrane-enclosed tubules and vesicles, extending from the nuclear membrane throughout the cytoplasm. ER is the largest organelle of most eukaryotic cells, as its membrane may account for at least 50% of all cell membranes and even more for specialized secretory cell. Its total area is 10–30 times that of the plasma membrane. ER constitutes the first compartment of the secretory pathway in which secreted and transmembrane proteins are folded and post-translationally modified [1].
ER is also the most important compartment for intracellular calcium ions (Ca2+) storage, which is necessary for the physiological activities of the ER, allowing the maintenance of the oxidation–reduction potential [2,3]. In its lumen, a set of specialized proteins like chaperones, foldases, glycosylating enzymes, oxidoreductases, and cofactors ensures the correct folding of newly synthesized proteins. By interacting with the exposed hydrophobic segments present on the newly synthesized proteins or on misfolded proteins, the chaperones (BiP/GRP78, calnexin, GRP94, etc.) act both to complete the folding process and to correct folding errors [4]. After passing the protein quality control checkpoints in the ER, correctly folded proteins traffic via the Golgi to other organelles and/or to the plasma membrane. Despite this optimized environment in the ER luminal domain, the success rate for accurate folding is variable. In case of unsuccessful folding, proteins are released in the cytosol where they become ubiquitinated and targeted to degradation by the proteasome. This rigorous quality control system has been named ERAD for Endoplasmic Reticulum-Associated Degradation [5].
In addition, to cope with the perturbations caused by unfolded or misfolded proteins, cells set off an adaptive response called the unfolded protein response (UPR), which aims to restore normal ER functioning [6,7,8,9]. This is achieved by (i) lowering the biosynthesis of proteins to reduce accumulation of misfolded proteins in the ER; (ii) increasing the biosynthesis of chaperone proteins; (iii) increasing ER size through membrane synthesis, (i), and (ii) resulting in a boost of ER folding capabilities; and finally (iv) increasing the biosynthesis of ER-associated degradation proteins thus improving the cell’s ability to eliminate misfolded proteins. Consequently, “adaptive UPR” limits cell damages and allows cell recovery and survival to a new stressful environment. However, if stress overcomes cell recovery capacities UPR can switch from an adaptive to a “terminal UPR” program triggering cell death [10,11,12].
Perturbations in the ER stress response such as either chronic ER stress or defects in UPR signaling, have been associated with a number of pathologies: diabetes, atherosclerosis, inflammation, stroke, pulmonary fibrosis, several eye diseases, neurodegenerative disorders (including amyotrophic lateral sclerosis, Alzheimer’s, Parkinson’s or Huntington’s diseases), and, of course, cancer [13,14,15,16]. The common feature among these seemingly different diseases is a cellular dysfunctioning leading to an accumulation of misfolded proteins in the ER.
With respect to cancer, the role of ER stress response/UPR signaling pathways was mainly studied in primary solid tumors in which a very unfavorable microenvironment mainly originating from inadequate vascularization and characterized by nutrient (e.g., amino acids, glucose) deprivation, hypoxia, acidosis leads to the activation of ER stress in the highly proliferative and metabolically active cancer cells [17,18,19,20,21]. However, in recent years our current knowledge on the essential functions played by the UPR in leukemia has also significantly improved.
In this review, after introducing the Unfolded Protein Response, we will summarize current findings on the involvement of ER stress in the progression of leukemia, and discuss the potential therapeutic effects of UPR activation or repression in these pathologies.

2. The Unfolded Protein Response

In mammals, UPR is triggered by activation of three ER transmembrane sensors: PERK (PKR-like ER-associated protein kinase), ATF6 (Activating Transcription Factor-6), and IRE1 (inositol-requiring enzyme-1) [6,10,22,23]. The luminal part of these proteins integrates the information coming from the ER lumen, whereas their cytosolic part interacts with their effectors and mediates the signaling cascades (Figure 1). In the absence of stress, the ER resident protein chaperone BiP also known as GRP78 (Glucose-regulated protein 78kDa) or HSPA5 (Heat Shock Protein Family A (Hsp70) Member 5) binds to the luminal domain of the three effectors and keep them in an inactive state. Upon accumulation of unfolded proteins in the ER lumen, BiP will act as a protein chaperone, interact with exposed hydrophobic segments of misfolded proteins, and thus be released from ATF6, IRE1, and PERK, leading to their activation [24,25]. In addition to BiP release, an activation of IRE1 by oligomerization induced by direct binding of unfolded proteins has also been reported, both in yeast [26] and mammalian cells [27]. Therefore, the relative ratios of three proteins complexes inside the endoplasmic reticulum, namely, those created by interaction between BiP and either unfolded proteins or UPR sensors, as well as those formed by direct interaction between unfolded proteins and the UPR sensors themselves, could contribute to a very precise and dynamic regulation of the UPR [28].
As previously stated, the primary goal of the activated signaling cascades is to reestablish ER homeostasis by a two-step process: in a first stage, through the reduction of overall protein synthesis and the degradation of misfolded proteins, and in a second stage through the activation of cellular functions crucial for cell survival [6,10,22].
However, in the absence of protein homeostasis restoration, the adaptive UPR will switch to terminal UPR, which ultimately results in cell death. Cell fate is largely influenced by the intensity and duration of the stress. A long or intense stress leads to the activation of this terminal UPR [10,18]. The regulatory networks, which determine the transition from adaptive to terminal UPR, are complex and not fully understood. Regardless, the molecular events that will direct the cell towards either adaptive or terminal UPR involve to some extent each of the PERK, IRE1, and ATF6 signaling cascades. The contribution of each pathway to the execution of the adaptive or terminal UPR may be variable depending on the type of cell and on the nature and extent of damage experienced by the cell. The different UPR signaling cascades are described below.

2.1. The Translational Pathway: Activation of the PERK Kinase

Among the three key proteins involved in UPR, PERK (encoded by the EIF2AK3 gene for eukaryotic translation initiation factor 2-alpha kinase 3) is the first to be activated by autophosphorylation. The dissociation of BiP from its luminal domain causes dimerization or oligomerization and trans-autophosphorylation of PERK (threonine 981), thus activating the cytosolic serine/threonine kinase domain (Figure 1). The main substrate of PERK is the alpha subunit of the translation initiation factor eIF2 (eukaryotic Initiation Factor 2) [29,30,31,32]. The eIF2 factor, which possesses three subunits-α, β, and γ-links the initiator methionine tRNA (tRNA-Met) to the small ribosomal subunit. The regulatory α subunit contains a serine (ser51) strictly conserved in eukaryotes. By phosphorylating ser51, PERK induces a global inhibition of cap-dependent translation initiation and therefore overall protein synthesis in order to temporary reduce unfolded protein load, until favorable conditions return [33]. Mechanistically, the phosphorylation of the eIF2α ser51 increases the affinity of eIF2 for its own eIF2B guanine nucleotide exchange factor (GEF), which recycles the inactive form of eIF2-GDP into its active form eIF2-GTP. This strong interaction induces sequestration of the eIF2 factor by eIF2B, causing a blockade of active translation pre-initiation complex formation and thus inhibition of translation initiation [34]. This translation inhibition prevents further protein loading in the ER, reduces cell overall metabolism and saves energy to repair the damage caused by the stress [34].
In parallel to cap-dependent translation arrest, translation of specific messenger RNAs exhibiting particular features in their 5′ untranslated region is selectively induced [35]. This is the case of the Activating Transcription Factor 4 (ATF4) mRNA which contains several upstream open frames (uORFs) in its 5′ untranslated region, preventing translation of the main open reading frame (ORF) in normal conditions. Under stress conditions however, low levels of active eIF2α allow the ribosomes to reach the main ATF4 ORF and efficiently initiate translation of this transcription factor, which in turn activates the expression of chaperones and of genes involved in amino acid metabolism and resistance to oxidative stress [36,37]. Interestingly, some mRNA whose translation depends on the presence of internal ribosome entry sites (IRESs) in their 5′ untranslated region [35] and coding for stress response proteins are also activated when eIF2α is phosphorylated (Figure 1) [38,39,40,41].
The dephosphorylation of eIF2α is necessary to restore a normal protein synthesis level after stress. This reset to the basal state is achieved by two phosphatases, composed of a single catalytic subunit PP1 (Protein Phosphatase 1) and one of the two regulatory subunits GADD34 (Growth And DNA-Damage inducible protein 34) or CReP (Constitutive Repressor of eIF2α phosphorylation) [42]. In contrast to CReP, which is constitutively expressed, the expression of GADD34 is only induced in response to stress as a negative feedback loop [43]. Indeed, transcription of GADD34 is activated by ATF4 and its translation is, as for ATF4 itself, regulated by a uORF mechanism ensuring proper GADD34 expression despite eIF2α phosphorylation [44].
Under chronic stress, sustained activation of PERK and thus prolonged expression of ATF4 induce apoptosis by activating CHOP transcription (C/EBP Homologous protein, also known as GADD153-Growth And DNA-Damage inducible protein 153 or DDIT3-DNA-Damage Inducible Transcript 3) [45]. This transcription factor, a member of the CCAAT/enhancer-binding protein (C/EBP) family, plays a central, multifunctional role in the UPR-induced apoptotic process [46]. CHOP can alone or cooperatively with other transcriptional factors function either as a transcriptional activator or repressor. It acts mainly by modulating the expression of various members of the BCL-2 protein family playing either pro-(Bim) or antiapoptotic (Bcl-2, BCL-XL and MCL-1) functions [47]. CHOP can however also induce cell death by many additional, non-exclusive, pathways such as restoration of protein synthesis (via GADD34 activation) which leads to increased proteins load detrimental to the cell (“proteotoxicity”) and by increased ROS production (through upregulation of the ER reductase ERO1α) [48].
It is interesting to note that eIF2α is not the only PERK substrate. Indeed, the transcription factor Nrf2 (Nuclear Factor (erythroid derived 2)-like2), which is involved in the response to oxidative stress, is normally maintained in the cytoplasm by association with Keap1. Under stress conditions, PERK phosphorylates Nrf2. This causes a dissociation of the Nrf2/Keap1 complex and allows the import of Nrf2 to nuclear compartment [49]. Nrf2 then bind to ARE sequences (Antioxidant response element) on the promoter of its target genes such as GCLC (Glutamate Cysteine Ligase Catalytic Subunit), HO-1 (Heme oxygenase 1) or NQO1 (NADPH dehydrogenase quinone 1) [50]. Thus, the activation of Nrf2 by PERK helps in maintaining the redox status of the cell subjected to ER stress.

2.2. The Transcriptional Pathway: Activation of ATF6α and IRE1α

In mammals, the transcriptional response to ER stress involves two families of transmembrane proteins: the IRE1 and ATF6 proteins (Figure 1).
The ATF6α (activating transcription factor 6 α) transcription factor is a type II transmembrane protein characterized by a C-terminal luminal domain, sensitive to misfolded proteins, and an N-terminal cytosolic portion containing a leucine zipper DNA binding domain (bZIP) and a transcriptional activation domain. In mammals, two ATF6 proteins, ATF6α and ATF6β, are produced form independent genes. Whereas both proteins are ubiquitously expressed, only ATF6α has proven to be an effective transcriptional activator and its it is currently accepted that only ATF6α plays a major role in the ATF6-dependent transduction of UPR signaling [51]. The amount and mode of contribution of ATF6β to the unfolded protein response remain poorly understood and need to be further investigated [52]. During ER stress, Bip dissociation from the ATF6α protein allows the exposure of two Golgi localization signals, and migration of ATF6α from ER to the Golgi apparatus where it undergoes 2 sequential cleavages by the proteins S1P and S2P (Site-1 and Site-2 Proteases) (Figure 1) [25,53]. These cleavages generate a transcriptionally active N-terminal short-lived fragment of 50 kDa called ATF6p50 which translocates into the nucleus to activate the transcription of chaperone and foldase proteins such as BiP, calreticulin, calnexin, and protein disulfide isomerases. ATF6p50 also activates the transcription of enzymes such as the calcium pump SERCA (sarco/endoplasmic reticulum Ca2+-ATPase). This ER ATPAse transports calcium ions from the cytosol into the ER and plays a major role in the maintenance of calcium homeostasis which controls many essential cellular processes [54]. ATF6p50 also promotes the expression of different genes involved in lipid biosynthesis, thus participating to the expansion of the endoplasmic reticulum [55]. It also upregulates XBP1 (X-box binding protein 1), a transcription factor which acts immediately downstream of the third UPR sensor IRE1 (see below). Moreover, ATF6α can also form heterodimers with XBP1 and upregulate genes involved in the ERAD pathway like EDEM (ER Degradation Enhancing Alpha-Mannosidase Like Protein 1) or HERPUD1 (Homocysteine Inducible ER Protein With Ubiquitin Like Domain 1). ATF6α gene invalidation induces increased sensitivity to ER stress probably due to impaired induction of chaperone proteins such as BiP or GRP94 (Glucose-regulated protein 94 kDa) [56,57]. However, ATF6α can also activate the expression of the proapoptotic factor CHOP [58,59], and a very recent work suggested that ATF6α could play an important role in the decision from adaptive to terminal UPR by modulating early and late CHOP expression kinetics [60]. Therefore, the role played by ATF6α on cell survival or death appears complex. In addition, the ATF6α transactivator domain (more precisely the first N-terminal 93 amino acids) has been shown to be responsible for its own degradation by the proteasome [61]. As a result, ATF6α appears as a powerful transcriptional activator, but with a transient effect. This may contribute to finely tune the UPR machinery.
The third UPR sensor is IRE1 (Inositol-requiring protein 1 also known as ERN1 for Endoplasmic reticulum-to-nucleus signaling 1), a 110 kDa protein initially identified in yeast where it is the only ER stress sensor. In mammals this protein is expressed as two isoforms: IRE1α, which is ubiquitously expressed, and IRE1β expressed only in the epithelial cells of the digestive system [62,63,64,65,66]. IRE1α possesses a luminal structure and an activation mode similar to that of PERK. However, in addition to a Ser/Thr kinase enzymatic activity, the IRE1α cytosolic domain also retains an atypical endoribonuclease (RNAse) activity, which becomes functional after IRE1α homodimerization under stress conditions [67]. This dimerization is essential for endoribonuclease activation, which is also dependent on IRE1α phosphorylation status [68]. The IRE1α RNAse domain catalyzes the excision of a 26-nucleotide sequence within the Xbp1 (X-box binding protein1) mRNA by an unconventional cytoplasmic splicing mechanism independent of the spliceosome (Figure 1) [69]. This cleavage, followed by a ligation step mediated by the RTCB tRNA ligase [70], generates a frame shift in the open reading frame, which leads to the expression of XBP1s (XBP1 spliced), a transcription factor belonging to the ATF/CREB family. The activation of the IRE1α/XBP1s signaling axis induces the expression of genes encoding proteins of the ERAD pathway (EDEM, HRD1) and factors that modulate protein translocation into the ER and folding, including the protein BiP [53,71]. Importantly, the non-spliced Xbp1 mRNA encodes the protein XBP1u (XBP1 unspliced), which is an inactive form with no transcriptional activity because it lacks the transactivating domain, and is an extremely short-lived protein. Interestingly, however, XBP1u was also found to interact with XBP1s under ER stress conditions, functioning as a negative feedback regulator [72,73].
IRE1α’s endoribonuclease activity has also been shown to induce rapid and specific degradation of some RNAs by a mechanism called RIDD (Regulated Ire1-Dependent Decay) (Figure 1) [74,75]. Currently, only a limited number of direct targets have been identified and validated, including 4 microRNAs (miR-17, 96, 125b, 34a) [76] and some mRNAs notably PER1 [77], SPARC [78], BLOS1 [79], and DR5 (death receptor 5), but bioinformatic studies coupled with transcriptomic studies suggest a wider spectrum of action [80,81]. Several studies indicate that the RIDD mechanism contributes to ER stress-induced cell death, notably by degrading several miRNAs involved in the repression of caspase-2 mRNA expression [75,76]. However, other studies propose that RIDD activity, by targeting mRNAs specifically translated at the endoplasmic reticulum, reduces the influx of newly synthesized proteins, and thus participates in the adaptive survival process [82]. Moreover, the IRE1α-mediated targeting through RIDD of the mRNA coding for the death receptor 5 protein, a cell surface transducer of apoptotic signals could also limit ER stress-induced cell death [83].
The IRE1α activation level, stability, conformation, and oligomerization status appear to be also regulated by the interaction with many different protein partners such as for example HSP47 which facilitates the dissociation of BiP from its luminal domain thus helping in activation of IRE1α signaling under low stress conditions [10,84,85].
IRE1α associates also with additional partners through its cytosolic domain to induce different signaling pathways. TRAF2 (TNF receptor-associated factor), an adaptor protein, associates with IRE1α’s kinase domain. The IRE1α/TRAF2 complex was found to interact with ASK1 (apoptosis signal-regulating kinase 1) to activate the c-Jun N-terminal kinase (JNK) and induce apoptosis [62,86]. Thus, the JNK arm of IRE1α pathway was initially thought to promote cell death. However, the function of this pathway in vivo is still controversial and has been described in some cases as pro-death and in other cases as pro-survival [62,86]. The nature and the intensity of the stimulus may account for these results. As in the case of ATF6α, IRE1α behaves as a sensor of the general cellular state through its multiple interactions with cofactors, regulators, and other members of the UPR signaling cascades and centralizes a set of signals in order to balance between anti- and proapoptotic signals.
Recent work has demonstrated that the activity of PERK and ATF6α can also be regulated by specific interacting proteins (reviewed in [10]). These results indicate that the activity of the three UPR effectors is extremely finely tuned. In addition, these effectors can establish crosstalk between each other during the UPR response and therefore more detailed analyses of these proteins and their identified partners remain necessary to better understand how they contribute on their own and altogether to the overall cell’s response during UPR activation.

3. Hematopoiesis and Leukemias

3.1. Hematopoiesis

Hematopoiesis is the physiological process that is responsible for the production of the mature pools of blood cells from undifferentiated precursors, the stem cells. Hematopoiesis, which takes place mainly in the bone marrow of long and flat bones, is a crucial process as it allows the maintenance of blood cell homeostasis, producing approximately 1012 blood cells daily in a healthy adult. The hematopoietic system functions as a pyramid-like hierarchy organized from a hematopoietic stem cell (HSC) at the top, able to self-renew or differentiate to produce all the cells of the hematopoietic system (Figure 2). In the bone marrow, long-term hematopoietic stem cells (LT-HSC) are quiescent, in the G0 phase of the cell cycle with a very low mitochondrial activity, but a high self-renewal potential [87]. These cells are maintained throughout life. In classical hematopoiesis, LT-HSC division leads to the generation of new LT-HSC or ST-HSC, for Short-Term Hematopoietic Stem Cell, which are able to produce all mature hematopoietic lineages [88,89]. These cells then differentiate into multipotent progenitors (MPPs) and then either in common lymphoid progenitors (CLPs) or in common myeloid progenitors (CMPs) [90]. CLPs produce B and T lymphocytes and natural killer cells, while CMPs generate granulocyte-macrophage progenitors (GMPs), which, as their name implies, are then differentiated into granulocytes and macrophages, and megakaryocyte–erythrocyte progenitors (MEPs) which themselves differentiate into red blood cells and platelets [91,92] (Figure 2).
Hematopoiesis is a tightly regulated mechanism, and therefore impaired hematopoiesis can be the cause of leukemias, malignant disorders resulting from defects of the stem cells at different stages of maturation, with subsequent clonal expansion [92]. Leukemias include acute and chronic leukemia and are also classified into lymphoblastic and myeloblastic leukemias according to the cell type affected. Acute leukemias are characterized by the proliferation of immature, unfunctional white blood cells called “blasts”, decreasing normal hematopoietic cells in the bone marrow while chronic leukemias are characterized by the expansion of differentiated cells in the blood [93]. Acute leukemias are divided into acute myeloid leukemias (AML) or acute lymphoblastic leukemias (ALL) and chronic leukemia into chronic myeloid leukemias (CML) or chronic lymphoblastic leukemias (CLL).

3.2. Acute Myeloid Leukemia (AML)

Acute myeloid leukemia (AML) is a group of phenotypically and genetically heterogeneous diseases, which is among the most common adult leukemia (it accounts for about 80% of leukemias in adults), with an average age of first diagnosis over 60 years [94]. This is a complex pathology triggered by the accumulation of chromosomal translocations and/or multiple mutations and resulting in the transformation and clonal expansion of hematopoietic progenitors. AML is thought to initially develop from at least two types of somatically acquired genetic alterations: mutations that confer advantages in terms of proliferation and survival and mutations that interfere with cell differentiation and apoptosis mechanisms [95]. Recent advances in sequencing methodologies have shown that AML represents a dynamic disorder in which multiple sub-clones compete and coexist, not only during the normal progression of the disease but also under pressure generated by anticancer agents [96]. While the majority of patients are in complete remission after the initial chemotherapy, AML has been associated with a poor prognosis because most patients tend to relapse due to the emergence of therapy-resistant clones [97]. Identifying the genetic alterations associated with resistance to chemotherapy is essential for risk stratification and to predict response to treatment of each AML patient. Three main classes of genetic aberrations have been described in AML: non-random chromosomal alterations, multiple gene mutations, and epigenetic alterations [98,99]. The most common of these chromosomal alterations include rearrangements leading to the formation of genes coding for chimeric proteins and upregulation of gene expression by juxtaposition with strong promoters. Among these rearrangements, we find translocations t(8;21) AML1-ETO or RUNX1, t(15;17) PML-RARA, inv(16) CBFB-MYH11, and t(9;11) MLL-AF9, which are associated with a better prognosis, whereas translocations t(11;19) MLL-ENL, t(6;11) MLL-AF6, t(10;11) MLL-AF10, or complex karyotypes are associated with a worse prognosis [100]. One of these translocations, t(15;17) (q22;q12), is peculiar because it is characteristic of a subtype of acute myeloid leukemia named acute promyelocytic leukemia (APL) [101]. This specific chromosomal translocation leads to the expression of the PML-RARα fusion protein. APL is unique among all leukemias because of its high level of sensitivity to all-trans retinoic acid (ATRA), the vitamin A acid form [102]. The prognosis of this pathology is very good in general [103,104]. Among the genes that have been found mutated in AML we can mention retinoic acid receptor-α (RAR-α), core binding factor (CBF), HOX gene family or MLL. Mutations in oncogenes such as FLT3, KIT, N-RAS, GATA-1, JUN B, MYC, p53, PU.1, RB, FES, FOS, MPL, WT1, WNT, CEBPA, and NPM1 or mutations affecting epigenetic modifiers such as DNMT3A, ASXL1, TET2, IDH1, and IDH2 have also been characterized [105,106,107].

3.3. Acute Lymphoblastic Leukemia (ALL)

Acute lymphoblastic leukemia (ALL), also called acute lymphocytic leukemia, is a rare genetically heterogeneous clonal malignant disorder of the bone marrow characterized by immature lymphoid precursors proliferation leading to the crowd out of normal hematopoietic cells [108]. ALL, which can occur at virtually any age, is more frequently seen in children and adolescents. This pathology results from clonal proliferation of abnormal B cell progenitors (B-ALL) accounting for approximately 85% of ALL or T cell progenitors (T-ALL) accounting for roughly 15% of ALL. Most of the genetic alterations leading to leukemogenesis, including chromosomal translocations, somatic mutations, aneuploidy, and gene copy number alterations have been characterized in both T-ALL and B-ALL. Like in AML, these genetic alterations are important prognostic factors for disease-risk stratification and treatment [109,110]. Among the genetic alterations found in B-ALL, TCF3–PBX1 t(1;19), ETV6–RUNX1 t(12;21), and hyperdiploidy are associated with a favorable outcome while MLL rearrangements, TCF3–HLF t(1;19) and rearrangements of CRLF2, JAK2A, or BL-class tyrosine kinase genes are of poor prognosis [111,112]. Alterations involving the KRAS, NRAS, FTL3, PTPN11, and epigenetic modifiers like CREBBP or WHSC1 are frequent genetic events [111,113].
The genetics of T-ALL is extremely heterogeneous, with chromosomal abnormalities in nearly all patients. Mutations in the NOTCH1 gene leading to constitutive activation of NOTCH signaling is the main oncogenic pathway found in the majority of patients. These alterations are generally associated with loss of p16 (INK4A) and p14 (ARF) suppressor genes at the CDKN2A locus. In addition, in 50% of patients with T-ALL, chromosomal translocations affect genes encoding oncogenic transcription factors like TAL1, TAL2, MYC, MYB, LYL1, TLX1 (HOX11), TLX3 (HOX11L2), or HOXA genes, placing these genes under the control of powerful T cell specific activators [114].
As for the AML example, it is not possible to exhaustively list all the genetic alterations and different combinations encountered, so we refer the reader to references dealing more specifically with this pathology [115,116,117].

3.4. Chronic Myeloid Leukemia (CML)

Chronic myeloid leukemia (CML) is a slow-growing myeloproliferative neoplasm characterized in more than 95% of cases by the t(9;22) (q34.1;q11.2) chromosomal translocation leading to the formation of the Philadelphia chromosome (Ph*), resulting in the BCR-ABL1 gene fusion. The subsequent BCR/ABL1 chimeric protein is a constitutively active tyrosine kinases oncoprotein which activates transduction pathways involved in cell growth and differentiation such as RAS, MYC, STAT, AKT RAF, or JUN, and is therefore capable of transforming hematopoietic stem cell into neoplastic one [118,119].
Before targeted therapies became available, the main treatment options for CML included allogeneic stem cell transplantation and chemotherapy. However, the prognosis for CML improved considerably since the use of tyrosine kinase inhibitors (TKIs), most notably imatinib in the early 2000s, which inhibit the BCR-ABL1 fusion protein by blocking its kinase domain [120]. Several generations of TKI have been developed since, but the appearance of TKI resistances remains a major issue [121]. It is therefore also crucial for this pathology to identify new therapeutic approaches in order to better stop its progression and avoid evolution to advanced disease states which may account for as much as 15% of all CML deaths [121].

3.5. Chronic Lymphocytic Leukemia (CLL)

Chronic lymphocytic leukemia (CLL) is characterized by a clonal proliferation and accumulation of mature but defective lymphocytes in the blood, bone marrow, lymph nodes, and spleen. CLL is the most common form of leukemia in Western countries. It is highly heterogeneous in its evolution, with some patients needing chemotherapy early after diagnosis and others never requiring specific treatment and having a survival rate similar to the general population. More than 95% of people with CLL develop the B cell type [122]. CLL is a heterogeneous disease, which divides into an aggressive form that expresses a wild type immunoglobulin heavy-chain variable region (IGVH) gene, and an indolent form that expresses a mutated IGVH, reflecting the stage of normal B cell differentiation [123,124]. Chronic lymphocytic leukemia cells exhibit many complex genetic alterations, which have been used by clinicians as prognostic biomarkers in order to predict survival and disease progression and guide treatment decisions [124]. Many recurrent cytogenetic abnormalities are encountered in CLL. The main ones are (i) deletion of the long arm of chromosome 13 (del(13q)), leading to the loss of the DLEU2/MIR15A/MIR16-1 genes, which is found in more than 50% of CLL cases and is of good prognosis when isolated; (ii) trisomy 12, associated with an intermediate prognosis with median overall survival; (iii) deletion of the long arm of chromosome 11 (del(11q)) that leads a more aggressive disease due to the loss of the ATM gene (for Ataxia Telangiectasia Mutated) which is essential for the regulation of the cell cycle; and (iv) deletion of the short arm of chromosome 17 (del(17p)) resulting in the loss of the TP53 gene which is of poor prognosis [125,126,127]. At least one of these abnormalities can be found in approximately 80% of patients [122,128,129]. Translocations are reported in approximately 20% of CLL [130]. These translocations predominantly involve the immunoglobulin genes, mainly IGH, and the 13q14 locus. Common partners are CCND1, BCL2, and BCL3 [130]. In addition to chromosomal rearrangements, sequencing studies have also revealed numerous recurrent mutations in CLL mostly in the P53, ATM, NOTCH1, SF3B1 (Splicing Factor 3B subunit 1), and BIRC3 genes [131].
A variety of targeted drugs including BCR signaling pathway inhibitors, anti-CD20 antibodies and BCL-2 inhibitors have been used in therapeutics and have significantly improved the management of this disease [132,133]. However, despite the increasing number of available therapeutic alternatives, chemotherapy does not currently provide a definitive cure and additional strategies are still required.

4. Endoplasmic Reticulum Stress Induction in Hematopoietic and Leukemic Cells

4.1. ER Stress Activation in HSCs

Hematopoietic stem cells (HSCs) are sitting at the apex of the hematopoietic hierarchy. They are the most immature cells and are capable of replenishing all hematopoietic cell types [134,135]. As long-life cells, HSCs require a highly regulated protein quality control in order to avoid the accumulation of damages that could ultimately affect their DNA integrity and promote tumorigenesis. At steady state, HSCs are quiescent and display lower protein synthesis rates in vivo and in vitro compared to their progeny [136]. Furthermore, HSCs have been associated with low protein folding capacity that can be explained by a lower expression of chaperones proteins compared to hematopoietic progenitors [137]. Moreover, protein synthesis deregulation has a great impact on HSCs’ viability and self-renewal capacities and can lead to HSCs loss [136,138]. Investigation of ER stress role in regulating hematopoietic stem cells fate, revealed a high expression of PERK and a low expression of eIF2α in HSCs when compared to progenitor cells [139,140]. PERK upregulation in HSCs appears to increase their sensitivity to ER stress, compared to more committed progenitor cells, through activation of the PERK-peIF2α-ATF4/CHOP arm that can trigger apoptosis. It has been suggested that this sensitivity to ER stress could prevent accumulation of damaged cells in the HSCs compartment and potential subsequent malignant transformation [139]. In agreement with this hypothesis, Miharada et al. showed that reducing ER stress levels in vitro in HSCs through the overexpression of the RNA binding protein Dppa5 (Developmental pluripotency-associated 5) improved their self-renewal activity by protecting them from apoptosis [141]. However, the IRE1α-XBP1 UPR branch can also be activated in HSCs and in this case plays a significant cytoprotective role. For example, estrogen treatment of HSCs activates the IRE1α-XBP1 branch and increases repopulation capacities of HSCs upon transplantation [142]. In a mouse model system, Liu et al. also showed that IRE1α-XBP1 activation in HSCs in vivo prevents ER stress-induced apoptosis, preserves HSC clonogenicity and improves reconstitution capacity [143]. Xie et al. also demonstrated that increased cytoprotective ER stress (induced by the pharmacological inhibition of the sphingolipid enzyme DEGS1) participates together with autophagy in the setting up of a prosurvival response aimed to maintain stemness properties [144].
Increased ERAD has also been recently reported to actively participate in the maintenance of proteins homeostasis in HSCs and appeared to be essential for stem cell pool maintenance [145]. In addition to low protein synthesis rates and low folding capacity, it has been reported that protein quality control by ERAD maintains HSCs pool. Altogether currently known data indicate that increased basal UPR induced at least in part by unfavorable growing conditions in the bone marrow environment, such as, e.g., hypoxia [146], helps in maintaining HSC integrity as well as clearing damaged HSCs and therefore play critical functions at the early steps of hematopoiesis [147]. Of note, in our article we refer to “basal UPR” as the activation status of the different signaling pathways of the UPR in cells growing either in vitro or in vivo without any treatment by chemotherapeutic drugs or chemical compounds.

4.2. ER Stress Activation in Leukemic Cells

Recent lines of evidence link activation of the three UPR branches to most hallmarks of cancer and especially those aimed to protect the cells against the numerous aggressions they undergo during their growth inside tumors [20]. This is especially true for solid tumors, which develop in a highly adverse environment but also for leukemic cells. Indeed, hematopoietic cells, either normal or leukemic, are exposed in the bone marrow to an adverse environment caused by hypoxia, high levels of reactive oxygen species (ROS), and nutrient deprivation, often resulting in ER stress activation [13,16,23,148]. Thus, many studies have reported the activation, to variable extents, of each of the three UPR branches (IRE1α, PERK, and ATF6α) in a wide range of hematopoietic tumors (leukemia, lymphoma, and myeloma) [137,149,150,151]. As in solid cancers, UPR plays a fundamental role in the adaptation of leukemic cells to cellular stress by inducing different mechanisms, which attempt to reestablish ER homeostasis in order to restore its proper functions.
In AML patients, increased expression of XBP1, BiP, and Calreticulin has been detected in 17.4% of cases [152]. Schardt et al. demonstrated a correlation between a high expression of XBP1s and complex karyotype in AML [152]. Another clinical study from Tanimura et al. reported activation of the IRE1α-XBP1 pathway in AML patients; however, no significant correlation between ER stress activation and genetic features could be revealed [153].
Interestingly, UPR activation in some hematological malignancies is not always the consequence of stress integration but can also be induced through aberrant pathway activation. For example, in chronic lymphoid leukemia (CLL), UPR activation is observed in response to surface immunoglobulin M stimulation and activation of the kinases BTK and SYK [154]. In pre-B-ALL, Xbp1 expression is activated by various oncogenic tyrosine kinases via STAT5 signaling [155]. Moreover, the transcription factor c-Jun, overexpressed in AML and CML, promotes the transcription of general UPR target genes such as Xbp1 and Atf4 by a direct mechanism [156]. The modulation of expression of some UPR effectors in leukemia has been shown to involve epigenetic modifications in their promoters [157,158].
In addition, mutations in epigenetic splicing factors, which are considered as first hit mutations, have pleiotropic effects that might be linked to ER stress activation (Figure 3). The comparison between healthy donor and AML patient samples revealed hypomethylation of Xbp1′s promoter that has been suggested to lead to overexpression of XBP1. On the contrary, in large diffuse B cells lymphoma, IRE1α expression is reduced through a mechanism involving the histone methyltransferase, EZH2 (Enhancer of Zeste Homolog 2).
Furthermore, transcription of ER stress-related proteins by oncogenic pathways also participates in the UPR activation in leukemia. For instance, the MAPK pathway promotes the transcription of Xbp1 through STAT5 activation. In pre-B acute lymphoblastic leukemia (ALL), BCR-ABL1 or NRASG12D signals through MAPK-STAT5-XBP1 [155]. Indirectly, in lymphomagenesis the transcription factor MYC, by promoting a rapid cell proliferation, increases the rate of misfolded proteins in ER that triggers the UPR [159].
Compared to HSC, in which both cytoprotective (IRE1α-XBP1) and cell death-promoting (PERK) UPR pathways can be activated in the basal state, the activation of an adaptive UPR (mainly via IRE1α signaling) appears to be preferred in leukemic cells. However, the increase in basal UPR could sensitize these cells to additional stress induced, for example, by chemotherapy treatment (see next chapter). By analogy with what is observed in HSCs under normal growth conditions, the UPR response may represent a real checkpoint influencing cell fate of leukemic cells experiencing chemotherapy: either the stress can be resolved via an adaptive phase and cancer progresses or the damage accumulates and becomes unrecoverable. In this latter case, excessive or prolonged stress triggers proapoptotic signaling through a terminal process [137,150,151,160]. This important issue is discussed below.

4.3. UPR Modulation: A Double-Edged Sword to Fight Against Leukemia

Despite the numerous pieces of evidence of reticulum stress activation in multiple cancers, the question of whether UPR reduces or promotes tumor growth in patients is still the subject of intense debate [149]. Two therapeutic strategies exploiting ER stress and UPR could be possible in order to induce leukemic cell death: either inhibition of the adaptive UPR response (cytoprotective) or activation of the terminal UPR response (cytotoxic). The choice between these two strategies may be difficult as their relative efficacy may be highly dependent on the cellular deregulation that led to the disease.
As mentioned above, various studies have shown that leukemic cells often possess basal UPR activity with a cytoprotective function, which favors tumor progression and additionally may increase chemoresistance of the cells to various drugs. For example, the ER stress sensor BiP was found to be highly expressed in B-ALL and its pharmacological inhibition by epigallocatechin gallate (a polyphenolic compound purified from green tea) sensitized cells to the anti-leukemic drug vincristine [161]. In the same pathology, the increase in expression and activity of BiP and the IRE1α/XBP1 pathway were found to be essential for cell survival and pharmacological inhibition of IRE1α RNAse domain by the drug STF-083010 reduced the proliferation and survival of patient-derived pre-B ALL cells [155]. Of note, increased Xbp1 mRNA levels at diagnosis appear of poor prognosis for patients with the disease [155]. In CML, activation of the PERK-eIF2α pathway has a cytoprotective effect and increases their resistance to imatinib, a tyrosine kinase inhibitor widely used in cancer chemotherapy [162]. Resistance to imatinib in CML was also shown to result from the activation of ATF6α, which appears mediated by the protein disulfide isomerase 5 (PDIA5) upon ER stress and a PDIA5 inhibitor, 16F16, increased cells’ sensitivity to treatment with imatinib [163]. Moreover, a pharmacological inhibitor of IRE1α, B109, was reported to suppress CLL tumor cell progression in a murine model and to sensitize human CLL cells to the Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib [164]. A pro-survival role for IRE1α was also reported in AML and the pharmacological inhibition of IRE1α by 2-hydroxy-1-naphthaldehyde (HNA) switched the cells towards apoptosis and in addition synergized with treatments with bortezomib and arsenic trioxide, two widely used anticancer drugs [157]. Moreover, analysis of Philadelphia chromosome (Ph)-positive AML patient samples revealed increased expression of the BiP, CHOP, and Xbp1s mRNAs and the authors demonstrated that inhibition of the IRE1α and ATF6α pathways sensitized cells expressing the Bcr-Abl fusion protein to imatinib- and etoposide-induced apoptosis [165]. More recently it was shown that Jun itself induces the expression of several UPR effectors thereby enhancing UPR induction and this appeared essential to AML cell proliferation and survival, thus demonstrating that Jun could contribute to induce an adaptive UPR in some AML subtypes [156].
Altogether, the data presented above have largely validated the inhibition of adaptive UPR as an effective means of fighting leukemia and a significant number of pharmacological inhibitors of central UPR effectors are currently under preclinical studies or clinical trials [137,150].
However, these promising results should not lead us to neglect the other strategy aimed at inducing a cytotoxic response in the cell through terminal UPR induction, the “second edge of the sword”. Indeed, leukemic cells, which usually experience unfavorable growth conditions and maintain increased levels of ER stress and basal UPR, may show an increased susceptibility to enter terminal UPR in response to different treatments. Indeed, artificially increasing the unfolded protein load can lead to a cytotoxic cellular response in some leukemic models. Thus, in ALL treatment with the drug pevonedistat, which inhibits the NEDD8 conjugation pathway and impairs degradation of misfolded proteins by the proteasome, induces a reorientation of UPR towards apoptosis [166]. Interestingly, inhibiting the ER-associated degradation (ERAD) pathway of proteins by knockdown of one of its components, UFD1, also results in the induction of a terminal UPR process in T-ALL cells in response to the accumulation of unfolded proteins [167]. In mast cell leukemia, it was demonstrated that moderate pharmacological inhibition of IRE1α could stop leukemic cell proliferation by impairing adaptive UPR but with non-significantly induced cell death. Interestingly, stronger inhibition of IRE1α induced a switch from adaptive to terminal UPR. Enhancing ER stress by pharmacological inhibition of proteasome activity with bortezomib also induced terminal UPR in this model [168]. In Philadelphia-positive ALL, pharmacological inhibition of IRE1α with MKC-8866 also appeared able to reorient the initial cytoprotective UPR program towards cell death induction when combined with the inhibition of BCR-ABL1 with nilotinib [169].
Few studies describing the induction of cell death in leukemic cells by a strategy deliberately aimed at redirecting the cell response towards terminal UPR has yet been described. However, the analysis of data published over the last two decades and describing the use of antileukemic drugs shows, strikingly, that for many of them (see Table 1) their mode of action involves the induction of a terminal UPR pathway or related UPR-induced cell death processes. Indeed, although adaptive UPR was found to contribute to chemoresistance in 10 out of the 91 chemical compounds tested against leukemic cells and listed in Table 1, for the remaining compounds (i.e., 89% of the whole) the induction of UPR signaling pathways was associated with cytotoxicity. There appears to be no apparent correlation between the type of leukemia and the final response, cytoprotective or cytotoxic, to UPR induction. This also seems to be the case if we consider the mode of action of the drugs used. Similarly, no strict correlation can be found between the UPR pathways activated in response to the drugs and the final response of the cell (pro-survival or pro-death) and all UPR pathway have been reported to be induced whatever the final outcome on leukemic cell’s viability. It can be noted, however, that the CHOP pathway is very frequently activated when UPR induction results in cell death. This appears not surprising as the PERK-peIF2α/ATF4/CHOP signaling pathway plays a crucial function in inducing cell apoptosis in the cell [46] and was reported to be a major cell death-inducing UPR pathway in hematopoietic stem cells, as described above (see Chapter 4.1). However, induction of the PERK-peIF2α/ATF4/CHOP signaling pathway can also be detected in leukemic cells responding to treatment by an adaptive UPR. As in other pathological models, leukemic cell response to UPR induction is a complex process, which may rely on a subtle balance between the activation levels of the different branches of the UPR. Anyway, it appears that for a large number of chemotherapeutic agents or candidate compound, this process is critical for the final death/survival outcome of leukemic cells.
Therefore, it seems important to further investigate terminal UPR induction, on its own as well as in combination with other pharmacological treatments, for the improvement of therapeutic strategies in leukemia.

5. Conclusions

We have reviewed the currently available data in the literature dealing with the various roles played by UPR in leukemia. We also presented some of the UPR-mediated molecular processes that can induce cytoprotection of leukemic cells or direct them towards cell death through apoptosis induction. In the light of all of the currently reported data from studies carried out to dissect the role of UPR in the progression of leukemia, it is clear that the cytoprotective/cytotoxic balance regulation is a complex, highly dynamic machinery, still poorly understood and that a wide and integrative approach is needed to discover the genuine mechanisms underlying this crucial process. The specific networks that regulate ER stress-induced cytoprotection or apoptosis may be dependent on the nature, the intensity and the length of the stimuli. It is probable, even if contradictory results have sometimes been published, that it also depends on the cell type being stressed.
A better understanding of the UPR mechanisms acting in response to chemotherapy appears also essential to provide new therapeutic pathways aimed to eradicate neoplastic cells either by inhibiting the adaptive UPR, or by activating UPR-mediated cell death pathways [265].

Funding

This work was supported the Institut National de la Santé et de la Recherche Médicale (INSERM), Université Toulouse III (Paul Sabatier) and Association Laurette Fugain (ALF2018/03 awarded to CT). KRP’s lab is funded by Kay Kendall Leukaemia Fund #KKL1149, Academy of Medical Sciences #SBF0041099 and Barts Charity. CP and MJ were supported by a fellowship from the French ministry of higher education and research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alberts, B.; Johnson, A.; Lewis, J.; Morgan, D.; Raff, M.; Roberts, K.; Walter, P. Molecular Biology of the Cell, 6th ed.; Garland Science: New York, NY, USA, 2014. [Google Scholar]
  2. Araki, K.; Nagata, K. Protein folding and quality control in the ER. Cold Spring Harb. Perspect. Biol. 2011, 3, a007526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Eletto, D.; Chevet, E.; Argon, Y.; Appenzeller-Herzog, C. Redox controls UPR to control redox. J. Cell Sci. 2014, 127, 3649–3658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sun, Z.; Brodsky, J.L. Protein quality control in the secretory pathway. J. Cell Biol. 2019, 218, 3171–3187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Li, C.; Xia, B.; Wang, S.; Xu, J. Folded or Degraded in Endoplasmic Reticulum. Adv. Exp. Med. Biol. 2020, 1248, 265–294. [Google Scholar] [CrossRef] [PubMed]
  6. Hetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102. [Google Scholar] [CrossRef]
  7. Ron, D.; Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 2007, 8, 519–529. [Google Scholar] [CrossRef]
  8. Moore, K.A.; Hollien, J. The unfolded protein response in secretory cell function. Annu. Rev. Genet. 2012, 46, 165–183. [Google Scholar] [CrossRef]
  9. Kaufman, R.J. Stress signaling from the lumen of the endoplasmic reticulum: Coordination of gene transcriptional and translational controls. Genes. Dev. 1999, 13, 1211–1233. [Google Scholar] [CrossRef] [Green Version]
  10. Hetz, C.; Zhang, K.; Kaufman, R.J. Mechanisms, regulation and functions of the unfolded protein response. Nat. Rev. Mol. Cell Biol. 2020, 21, 421–438. [Google Scholar] [CrossRef]
  11. Sicari, D.; Igbaria, A.; Chevet, E. Control of Protein Homeostasis in the Early Secretory Pathway: Current Status and Challenges. Cells 2019, 8, 1347. [Google Scholar] [CrossRef] [Green Version]
  12. Maurel, M.; McGrath, E.P.; Mnich, K.; Healy, S.; Chevet, E.; Samali, A. Controlling the unfolded protein response-mediated life and death decisions in cancer. Semin. Cancer Biol. 2015, 33, 57–66. [Google Scholar] [CrossRef] [PubMed]
  13. Hetz, C.; Chevet, E. Theme Series—UPR in cancer. Semin. Cancer Biol. 2015, 33, 1–2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hosoi, T.; Ozawa, K. Endoplasmic reticulum stress in disease: Mechanisms and therapeutic opportunities. Clin. Sci. 2009, 118, 19–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hetz, C.; Axten, J.M. Pharmacological targeting of the unfolded protein response for disease intervention. Nat. Chem. Biol. 2019, 15, 764–775. [Google Scholar] [CrossRef] [PubMed]
  16. Hetz, C.; Chevet, E.; Harding, H.P. Targeting the unfolded protein response in disease. Nat. Rev. Drug Discov. 2013, 12, 703–719. [Google Scholar] [CrossRef] [PubMed]
  17. Corazzari, M.; Gagliardi, M.; Fimia, G.M.; Piacentini, M. Endoplasmic Reticulum Stress, Unfolded Protein Response, and Cancer Cell Fate. Nat. Chem. Biol. 2017, 7, 78. [Google Scholar] [CrossRef] [Green Version]
  18. Oakes, S.A. Endoplasmic Reticulum Stress Signaling in Cancer Cells. Am. J. Pathol. 2020, 190, 934–946. [Google Scholar] [CrossRef] [Green Version]
  19. Oakes, S.A. Endoplasmic reticulum proteostasis: A key checkpoint in cancer. Am. J. Physiol. Cell Physiol. 2017, 312, C93–C102. [Google Scholar] [CrossRef] [Green Version]
  20. Urra, H.; Dufey, E.; Avril, T.; Chevet, E.; Hetz, C. Endoplasmic Reticulum Stress and the Hallmarks of Cancer. Trends Cancer 2016, 2, 252–262. [Google Scholar] [CrossRef]
  21. Chevet, E.; Hetz, C.; Samali, A. Endoplasmic reticulum stress-activated cell reprogramming in oncogenesis. Cancer Discov. 2015, 5, 586–597. [Google Scholar] [CrossRef] [Green Version]
  22. Korennykh, A.; Walter, P. Structural basis of the unfolded protein response. Annu. Rev. Cell Dev. Biol. 2012, 28, 251–277. [Google Scholar] [CrossRef] [PubMed]
  23. Hetz, C.; Papa, F.R. The Unfolded Protein Response and Cell Fate Control. Mol. Cell 2018, 69, 169–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Bertolotti, A.; Zhang, Y.; Hendershot, L.M.; Harding, H.P.; Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat. Cell Biol. 2000, 2, 326–332. [Google Scholar] [CrossRef] [PubMed]
  25. Shen, J.; Chen, X.; Hendershot, L.; Prywes, R. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev. Cell 2002, 3, 99–111. [Google Scholar] [CrossRef] [Green Version]
  26. Gardner, B.M.; Walter, P. Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response. Science 2011, 333, 1891–1894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Karagoz, G.E.; Acosta-Alvear, D.; Nguyen, H.T.; Lee, C.P.; Chu, F.; Walter, P. An unfolded protein-induced conformational switch activates mammalian IRE1. eLife 2017, 6, e30700. [Google Scholar] [CrossRef] [PubMed]
  28. Bakunts, A.; Orsi, A.; Vitale, M.; Cattaneo, A.; Lari, F.; Tade, L.; Sitia, R.; Raimondi, A.; Bachi, A.; van Anken, E. Ratiometric sensing of BiP-client versus BiP levels by the unfolded protein response determines its signaling amplitude. eLife 2017, 6, e27518. [Google Scholar] [CrossRef]
  29. Harding, H.P.; Zhang, Y.; Ron, D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 1999, 397, 271–274. [Google Scholar] [CrossRef]
  30. Harding, H.P.; Novoa, I.; Zhang, Y.; Zeng, H.; Wek, R.; Schapira, M.; Ron, D. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 2000, 6, 1099–1108. [Google Scholar] [CrossRef]
  31. Marciniak, S.J.; Garcia-Bonilla, L.; Hu, J.; Harding, H.P.; Ron, D. Activation-dependent substrate recruitment by the eukaryotic translation initiation factor 2 kinase PERK. J. Cell Biol. 2006, 172, 201–209. [Google Scholar] [CrossRef]
  32. DuRose, J.B.; Scheuner, D.; Kaufman, R.J.; Rothblum, L.I.; Niwa, M. Phosphorylation of eukaryotic translation initiation factor 2alpha coordinates rRNA transcription and translation inhibition during endoplasmic reticulum stress. Mol. Cell. Biol. 2009, 29, 4295–4307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Koumenis, C.; Naczki, C.; Koritzinsky, M.; Rastani, S.; Diehl, A.; Sonenberg, N.; Koromilas, A.; Wouters, B.G. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha. Mol. Cell. Biol. 2002, 22, 7405–7416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wek, R.C.; Cavener, D.R. Translational control and the unfolded protein response. Antioxid. Redox Signal. 2007, 9, 2357–2371. [Google Scholar] [CrossRef] [PubMed]
  35. Jaud, M.; Philippe, C.; Di Bella, D.; Tang, W.; Pyronnet, S.; Laurell, H.; Mazzolini, L.; Rouault-Pierre, K.; Touriol, C. Translational Regulations in Response to Endoplasmic Reticulum Stress in Cancers. Cells 2020, 9, 540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Vattem, K.M.; Wek, R.C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc. Natl. Acad. Sci. USA 2004, 101, 11269–11274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Palam, L.R.; Baird, T.D.; Wek, R.C. Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation. J. Biol. Chem. 2011, 286, 10939–10949. [Google Scholar] [CrossRef] [Green Version]
  38. Bonnet-Magnaval, F.; Philippe, C.; Van Den Berghe, L.; Prats, H.; Touriol, C.; Lacazette, E. Hypoxia and ER stress promote Staufen1 expression through an alternative translation mechanism. Cancers 2016, 479, 365–371. [Google Scholar] [CrossRef]
  39. Jaud, M.; Philippe, C.; Van Den Berghe, L.; Ségura, C.; Mazzolini, L. The PERK Branch of the Unfolded Protein Response Promotes DLL4 Expression by Activating an Alternative Translation Mechanism. Cancers 2019, 11, 142. [Google Scholar] [CrossRef] [Green Version]
  40. Philippe, C.; Dubrac, A.; Quelen, C.; Desquesnes, A.; Van Den Berghe, L.; Ségura, C.; Filleron, T.; Pyronnet, S.; Prats, H.; Brousset, P.; et al. PERK mediates the IRES-dependent translational activation of mRNAs encoding angiogenic growth factors after ischemic stress. Sci. Signal. 2016, 9, ra44. [Google Scholar] [CrossRef]
  41. Fernandez, J.; Bode, B.; Koromilas, A.; Diehl, J.A.; Krukovets, I.; Snider, M.D.; Hatzoglou, M. Translation mediated by the internal ribosome entry site of the cat-1 mRNA is regulated by glucose availability in a PERK kinase-dependent manner. J. Biol. Chem. 2002, 277, 11780–11787. [Google Scholar] [CrossRef] [Green Version]
  42. Reid, D.W.; Tay, A.S.; Sundaram, J.R.; Lee, I.C.; Chen, Q.; George, S.E.; Nicchitta, C.V.; Shenolikar, S. Complementary Roles of GADD34- and CReP-Containing Eukaryotic Initiation Factor 2α Phosphatases during the Unfolded Protein Response. Mol. Cell. Biol. 2016, 36, 1868–1880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ma, Y.; Hendershot, L.M. Delineation of a negative feedback regulatory loop that controls protein translation during endoplasmic reticulum stress. J. Biol. Chem. 2003, 278, 34864–34873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Lee, Y.Y.; Cevallos, R.C.; Jan, E. An upstream open reading frame regulates translation of GADD34 during cellular stresses that induce eIF2alpha phosphorylation. J. Biol. Chem. 2009, 284, 6661–6673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ma, Y.; Brewer, J.W.; Diehl, J.A.; Hendershot, L.M. Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J. Mol. Biol. 2002, 318, 1351–1365. [Google Scholar] [CrossRef]
  46. 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. 2018, 9, 3083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Rozpedek, W.; Pytel, D.; Mucha, B.; Leszczynska, H.; Diehl, J.A.; Majsterek, I. The Role of the PERK/eIF2α/ATF4/CHOP Signaling Pathway in Tumor Progression During Endoplasmic Reticulum Stress. Curr. Mol. Med. 2016, 16, 533–544. [Google Scholar] [CrossRef] [PubMed]
  48. Marciniak, S.J.; Yun, C.Y.; Oyadomari, S.; Novoa, I.; Zhang, Y.; Jungreis, R.; Nagata, K.; Harding, H.P.; Ron, D. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes. Dev. 2004, 18, 3066–3077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Cullinan, S.B.; Zhang, D.; Hannink, M.; Arvisais, E.; Kaufman, R.J.; Diehl, J.A. Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol. Cell. Biol. 2003, 23, 7198–7209. [Google Scholar] [CrossRef] [Green Version]
  50. Cullinan, S.B.; Diehl, J.A. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J. Biol. Chem. 2004, 279, 20108–20117. [Google Scholar] [CrossRef] [Green Version]
  51. Bobrovnikova-Marjon, E.; Diehl, J.A. Coping with stress: ATF6alpha takes the stage. Dev. Cell 2007, 13, 322–324. [Google Scholar] [CrossRef] [Green Version]
  52. Hillary, R.F.; FitzGerald, U. A lifetime of stress: ATF6 in development and homeostasis. J. Biomed. Sci. 2018, 25, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yamamoto, K.; Sato, T.; Matsui, T.; Sato, M.; Okada, T.; Yoshida, H.; Harada, A.; Mori, K. Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1. Dev. Cell 2007, 13, 365–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chemaly, E.R.; Troncone, L.; Lebeche, D. SERCA control of cell death and survival. Cell Calcium 2018, 69, 46–61. [Google Scholar] [CrossRef] [PubMed]
  55. Bommiasamy, H.; Back, S.H.; Fagone, P.; Lee, K.; Meshinchi, S.; Vink, E.; Sriburi, R.; Frank, M.; Jackowski, S.; Kaufman, R.J.; et al. ATF6alpha induces XBP1-independent expansion of the endoplasmic reticulum. J. Cell Sci. 2009, 122, 1626–1636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Yamamoto, K.; Takahara, K.; Oyadomari, S.; Okada, T.; Sato, T.; Harada, A.; Mori, K. Induction of liver steatosis and lipid droplet formation in ATF6alpha-knockout mice burdened with pharmacological endoplasmic reticulum stress. Mol. Biol. Cell 2010, 21, 2975–2986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wu, J.; Rutkowski, D.T.; Dubois, M.; Swathirajan, J.; Saunders, T.; Wang, J.; Song, B.; Yau, G.D.; Kaufman, R.J. ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress. Dev. Cell 2007, 13, 351–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Guan, D.; Xu, Y.; Yang, M.; Wang, H.; Wang, X.; Shen, Z. N-acetyl cysteine and penicillamine induce apoptosis via the ER stress response-signaling pathway. Mol. Carcinog. 2010, 49, 68–74. [Google Scholar] [CrossRef]
  59. Yoshida, H.; Okada, T.; Haze, K.; Yanagi, H.; Yura, T.; Negishi, M.; Mori, K. ATF6 activated by proteolysis binds in the presence of NF-Y (CBF) directly to the cis-acting element responsible for the mammalian unfolded protein response. Mol. Cell. Biol. 2000, 20, 6755–6767. [Google Scholar] [CrossRef] [Green Version]
  60. Yang, H.; Niemeijer, M.; van de Water, B.; Beltman, J.B. ATF6 Is a Critical Determinant of CHOP Dynamics during the Unfolded Protein Response. iScience 2020, 23, 100860. [Google Scholar] [CrossRef] [Green Version]
  61. Thuerauf, D.J.; Morrison, L.E.; Hoover, H.; Glembotski, C.C. Coordination of ATF6-mediated transcription and ATF6 degradation by a domain that is shared with the viral transcription factor, VP16. J. Biol. Chem. 2002, 277, 20734–20739. [Google Scholar] [CrossRef] [Green Version]
  62. Urano, F.; Bertolotti, A.; Ron, D. IRE1 and efferent signaling from the endoplasmic reticulum. J. Cell Sci. 2000, 113 Pt 21, 3697–3702. [Google Scholar]
  63. Tirasophon, W.; Welihinda, A.A.; Kaufman, R.J. A stress response pathway from the endoplasmic reticulum to the nucleus requires a novel bifunctional protein kinase/endoribonuclease (Ire1p) in mammalian cells. Genes. Dev. 1998, 12, 1812–1824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wang, X.Z.; Harding, H.P.; Zhang, Y.; Jolicoeur, E.M.; Kuroda, M.; Ron, D. Cloning of mammalian Ire1 reveals diversity in the ER stress responses. EMBO J. 1998, 17, 5708–5717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bertolotti, A.; Wang, X.; Novoa, I.; Jungreis, R.; Schlessinger, K.; Cho, J.H.; West, A.B.; Ron, D. Increased sensitivity to dextran sodium sulfate colitis in IRE1beta-deficient mice. J. Clin. Investig. 2001, 107, 585–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Martino, M.B.; Jones, L.; Brighton, B.; Ehre, C.; Abdulah, L.; Davis, C.W.; Ron, D.; O’Neal, W.K.; Ribeiro, C.M. The ER stress transducer IRE1beta is required for airway epithelial mucin production. Mucosal Immunol. 2013, 6, 639–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Shamu, C.E.; Walter, P. Oligomerization and phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus. EMBO J. 1996, 15, 3028–3039. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, C.Y.; Schröder, M.; Kaufman, R.J. Ligand-independent dimerization activates the stress response kinases IRE1 and PERK in the lumen of the endoplasmic reticulum. J. Biol. Chem. 2000, 275, 24881–24885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Peschek, J.; Acosta-Alvear, D.; Mendez, A.S.; Walter, P. A conformational RNA zipper promotes intron ejection during non-conventional XBP1 mRNA splicing. EMBO Rep. 2015, 16, 1688–1698. [Google Scholar] [CrossRef]
  70. Lu, Y.; Liang, F.X.; Wang, X. A synthetic biology approach identifies the mammalian UPR RNA ligase RtcB. Mol. Cell 2014, 55, 758–770. [Google Scholar] [CrossRef] [Green Version]
  71. Acosta-Alvear, D.; Zhou, Y.; Blais, A.; Tsikitis, M.; Lents, N.H.; Arias, C.; Lennon, C.J.; Kluger, Y.; Dynlacht, B.D. XBP1 controls diverse cell type- and condition-specific transcriptional regulatory networks. Mol. Cell 2007, 27, 53–66. [Google Scholar] [CrossRef]
  72. Guo, F.; Lin, E.A.; Liu, P.; Lin, J.; Liu, C. XBP1U inhibits the XBP1S-mediated upregulation of the iNOS gene expression in mammalian ER stress response. Cell. Signal. 2010, 22, 1818–1828. [Google Scholar] [CrossRef] [PubMed]
  73. Byrd, A.E.; Brewer, J.W. Intricately Regulated: A Cellular Toolbox for Fine-Tuning XBP1 Expression and Activity. Cells 2012, 1, 738–753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hollien, J.; Weissman, J.S. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science 2006, 313, 104–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Maurel, M.; Chevet, E.; Tavernier, J.; Gerlo, S. Getting RIDD of RNA: IRE1 in cell fate regulation. Trends Biochem. Sci. 2014, 39, 245–254. [Google Scholar] [CrossRef] [PubMed]
  76. Upton, J.P.; Wang, L.; Han, D.; Wang, E.S.; Huskey, N.E.; Lim, L.; Truitt, M.; McManus, M.T.; Ruggero, D.; Goga, A.; et al. IRE1alpha cleaves select microRNAs during ER stress to derepress translation of proapoptotic Caspase-2. Science 2012, 338, 818–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Pluquet, O.; Dejeans, N.; Bouchecareilh, M.; Lhomond, S.; Pineau, R.; Higa, A.; Delugin, M.; Combe, C.; Loriot, S.; Cubel, G.; et al. Posttranscriptional regulation of PER1 underlies the oncogenic function of IREalpha. Cancer Res. 2013, 73, 4732–4743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Dejeans, N.; Pluquet, O.; Lhomond, S.; Grise, F.; Bouchecareilh, M.; Juin, A.; Meynard-Cadars, M.; Bidaud-Meynard, A.; Gentil, C.; Moreau, V.; et al. Autocrine control of glioma cells adhesion and migration through IRE1alpha-mediated cleavage of SPARC mRNA. J. Cell Sci. 2012, 125, 4278–4287. [Google Scholar] [CrossRef] [Green Version]
  79. Bright, M.D.; Itzhak, D.N.; Wardell, C.P.; Morgan, G.J.; Davies, F.E. Cleavage of BLOC1S1 mRNA by IRE1 Is Sequence Specific, Temporally Separate from XBP1 Splicing, and Dispensable for Cell Viability under Acute Endoplasmic Reticulum Stress. Mol. Cell. Biol. 2015, 35, 2186–2202. [Google Scholar] [CrossRef] [Green Version]
  80. Han, D.; Lerner, A.G.; Vande Walle, L.; Upton, J.P.; Xu, W.; Hagen, A.; Backes, B.J.; Oakes, S.A.; Papa, F.R. IRE1alpha kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell 2009, 138, 562–575. [Google Scholar] [CrossRef] [Green Version]
  81. So, J.S.; Hur, K.Y.; Tarrio, M.; Ruda, V.; Frank-Kamenetsky, M.; Fitzgerald, K.; Koteliansky, V.; Lichtman, A.H.; Iwawaki, T.; Glimcher, L.H.; et al. Silencing of lipid metabolism genes through IRE1alpha-mediated mRNA decay lowers plasma lipids in mice. Cell Metab. 2012, 16, 487–499. [Google Scholar] [CrossRef] [Green Version]
  82. Moore, K.; Hollien, J. Ire1-mediated decay in mammalian cells relies on mRNA sequence, structure, and translational status. Mol. Biol. Cell 2015, 26, 2873–2884. [Google Scholar] [CrossRef] [PubMed]
  83. Lu, M.; Lawrence, D.A.; Marsters, S.; Acosta-Alvear, D.; Kimmig, P.; Mendez, A.S.; Paton, A.W.; Paton, J.C.; Walter, P.; Ashkenazi, A. Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science 2014, 345, 98–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hetz, C.; Glimcher, L.H. Fine-tuning of the unfolded protein response: Assembling the IRE1alpha interactome. Mol. Cell 2009, 35, 551–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sepulveda, D.; Rojas-Rivera, D.; Rodriguez, D.A.; Groenendyk, J.; Kohler, 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 IRE1alpha. Mol. Cell 2018, 69, 238–252.e237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Urano, F.; Wang, X.; Bertolotti, A.; Zhang, Y.; Chung, P.; Harding, H.P.; Ron, D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000, 287, 664–666. [Google Scholar] [CrossRef] [Green Version]
  87. Zhang, Y.; Gao, S.; Xia, J.; Liu, F. Hematopoietic Hierarchy—An Updated Roadmap. Trends Cell Biol. 2018, 28, 976–986. [Google Scholar] [CrossRef]
  88. Chao, M.P.; Seita, J.; Weissman, I.L. Establishment of a normal hematopoietic and leukemia stem cell hierarchy. Cold Spring Harb. Symp. Quant. Biol. 2008, 73, 439–449. [Google Scholar] [CrossRef]
  89. Warr, M.R.; Pietras, E.M.; Passegué, E. Mechanisms controlling hematopoietic stem cell functions during normal hematopoiesis and hematological malignancies. Wiley Interdiscip. Rev. Syst. Biol. Med. 2011, 3, 681–701. [Google Scholar] [CrossRef]
  90. Noetzli, L.J.; French, S.L.; Machlus, K.R. New Insights into the Differentiation of Megakaryocytes from Hematopoietic Progenitors. Arter. Thromb. Vasc. Biol. 2019, 39, 1288–1300. [Google Scholar] [CrossRef]
  91. Akashi, K.; Traver, D.; Miyamoto, T.; Weissman, I.L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 2000, 404, 193–197. [Google Scholar] [CrossRef]
  92. Passegué, E.; Jamieson, C.H.; Ailles, L.E.; Weissman, I.L. Normal and leukemic hematopoiesis: Are leukemias a stem cell disorder or a reacquisition of stem cell characteristics? Proc. Natl. Acad. Sci. USA 2003, 100 (Suppl. 1), 11842–11849. [Google Scholar] [CrossRef] [Green Version]
  93. Sawyers, C.L.; Denny, C.T.; Witte, O.N. Leukemia and the disruption of normal hematopoiesis. Cell 1991, 64, 337–350. [Google Scholar] [CrossRef]
  94. De Kouchkovsky, I.; Abdul-Hay, M. Acute myeloid leukemia: A comprehensive review and 2016 update. Blood Cancer J. 2016, 6, e441. [Google Scholar] [CrossRef] [PubMed]
  95. Grove, C.S.; Vassiliou, G.S. Acute myeloid leukaemia: A paradigm for the clonal evolution of cancer? Dis. Models Mech. 2014, 7, 941–951. [Google Scholar] [CrossRef] [Green Version]
  96. Klco, J.M.; Spencer, D.H.; Miller, C.A.; Griffith, M.; Lamprecht, T.L.; O’Laughlin, M.; Fronick, C.; Magrini, V.; Demeter, R.T.; Fulton, R.S.; et al. Functional heterogeneity of genetically defined subclones in acute myeloid leukemia. Cancer Cell 2014, 25, 379–392. [Google Scholar] [CrossRef] [Green Version]
  97. Hackl, H.; Astanina, K.; Wieser, R. Molecular and genetic alterations associated with therapy resistance and relapse of acute myeloid leukemia. J. Hematol. Oncol. 2017, 10, 51. [Google Scholar] [CrossRef] [Green Version]
  98. Lagunas-Rangel, F.A.; Chávez-Valencia, V.; Gómez-Guijosa, M.; Cortes-Penagos, C. Acute Myeloid Leukemia-Genetic Alterations and Their Clinical Prognosis. Int. J. Hematol. Oncol. Stem Cell Res. 2017, 11, 328–339. [Google Scholar]
  99. Goldman, S.L.; Hassan, C.; Khunte, M.; Soldatenko, A.; Jong, Y.; Afshinnekoo, E.; Mason, C.E. Epigenetic Modifications in Acute Myeloid Leukemia: Prognosis, Treatment, and Heterogeneity. Front. Genet. 2019, 10, 133. [Google Scholar] [CrossRef] [Green Version]
  100. Pourrajab, F.; Zare-Khormizi, M.R.; Hashemi, A.S.; Hekmatimoghaddam, S. Genetic Characterization and Risk Stratification of Acute Myeloid Leukemia. Cancer Manag. Res. 2020, 12, 2231–2253. [Google Scholar] [CrossRef] [Green Version]
  101. Benedetti, L.; Levin, A.A.; Scicchitano, B.M.; Grignani, F.; Allenby, G.; Diverio, D.; Lo Coco, F.; Avvisati, G.; Ruthardt, M.; Adamo, S.; et al. Characterization of the retinoid binding properties of the major fusion products present in acute promyelocytic leukemia cells. Blood 1997, 90, 1175–1185. [Google Scholar] [CrossRef]
  102. Visani, G.; Buonamici, S.; Malagola, M.; Isidori, A.; Piccaluga, P.P.; Martinelli, G.; Ottaviani, E.; Grafone, T.; Baccarani, M.; Tura, S. Pulsed ATRA as single therapy restores long-term remission in PML-RARalpha-positive acute promyelocytic leukemia patients: Real time quantification of minimal residual disease. A pilot study. Leukemia 2001, 15, 1696–1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Cicconi, L.; Divona, M.; Ciardi, C.; Ottone, T.; Ferrantini, A.; Lavorgna, S.; Alfonso, V.; Paoloni, F.; Piciocchi, A.; Avvisati, G.; et al. PML-RARα kinetics and impact of FLT3-ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy. Leukemia 2016, 30, 1987–1992. [Google Scholar] [CrossRef] [PubMed]
  104. Martinelli, G.; Ottaviani, E.; Visani, G.; Testoni, N.; Montefusco, V.; Tura, S. Long-term disease-free acute promyelocytic leukemia patients really can be cured at molecular level. Haematologica 1998, 83, 860–863. [Google Scholar] [PubMed]
  105. Haferlach, T.; Bacher, U.; Haferlach, C.; Kern, W.; Schnittger, S. Insight into the molecular pathogenesis of myeloid malignancies. Curr. Opin. Hematol. 2007, 14, 90–97. [Google Scholar] [CrossRef]
  106. Haferlach, T.; Kohlmann, A.; Bacher, U.; Schnittger, S.; Haferlach, C.; Kern, W. Gene expression profiling for the diagnosis of acute leukaemia. Br. J. Cancer 2007, 96, 535–540. [Google Scholar] [CrossRef] [Green Version]
  107. Prada-Arismendy, J.; Arroyave, J.C.; Röthlisberger, S. Molecular biomarkers in acute myeloid leukemia. Blood Rev. 2017, 31, 63–76. [Google Scholar] [CrossRef]
  108. Inaba, H.; Greaves, M.; Mullighan, C.G. Acute lymphoblastic leukaemia. Lancet 2013, 381, 1943–1955. [Google Scholar] [CrossRef] [Green Version]
  109. Teitell, M.A.; Pandolfi, P.P. Molecular genetics of acute lymphoblastic leukemia. Annu. Rev. Pathol. 2009, 4, 175–198. [Google Scholar] [CrossRef]
  110. Armstrong, S.A.; Look, A.T. Molecular genetics of acute lymphoblastic leukemia. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 6306–6315. [Google Scholar] [CrossRef]
  111. Moorman, A.V. New and emerging prognostic and predictive genetic biomarkers in B-cell precursor acute lymphoblastic leukemia. Haematologica 2016, 101, 407–416. [Google Scholar] [CrossRef] [Green Version]
  112. Palmi, C.; Savino, A.M.; Silvestri, D.; Bronzini, I.; Cario, G.; Paganin, M.; Buldini, B.; Galbiati, M.; Muckenthaler, M.U.; Bugarin, C.; et al. CRLF2 over-expression is a poor prognostic marker in children with high risk T-cell acute lymphoblastic leukemia. Oncotarget 2016, 7, 59260–59272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Malinowska-Ozdowy, K.; Frech, C.; Schönegger, A.; Eckert, C.; Cazzaniga, G.; Stanulla, M.; zur Stadt, U.; Mecklenbräuker, A.; Schuster, M.; Kneidinger, D.; et al. KRAS and CREBBP mutations: A relapse-linked malicious liaison in childhood high hyperdiploid acute lymphoblastic leukemia. Leukemia 2015, 29, 1656–1667. [Google Scholar] [CrossRef] [PubMed]
  114. Van Vlierberghe, P.; Ferrando, A. The molecular basis of T cell acute lymphoblastic leukemia. J. Clin. Investig. 2012, 122, 3398–3406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Malard, F.; Mohty, M. Acute lymphoblastic leukaemia. Lancet 2020, 395, 1146–1162. [Google Scholar] [CrossRef]
  116. Brown, L.M.; Lonsdale, A.; Zhu, A.; Davidson, N.M.; Schmidt, B.; Hawkins, A.; Wallach, E.; Martin, M.; Mechinaud, F.M.; Khaw, S.L.; et al. The application of RNA sequencing for the diagnosis and genomic classification of pediatric acute lymphoblastic leukemia. Blood Adv. 2020, 4, 930–942. [Google Scholar] [CrossRef] [PubMed]
  117. Waanders, E.; Gu, Z.; Dobson, S.M.; Antić, Ž.; Crawford, J.C.; Ma, X.; Edmonson, M.N.; Payne-Turner, D.; van der Vorst, M.; Jongmans, M.C.J.; et al. Mutational landscape and patterns of clonal evolution in relapsed pediatric acute lymphoblastic leukemia. Blood Cancer Discov. 2020, 1, 96–111. [Google Scholar] [CrossRef]
  118. Apperley, J.F. Chronic myeloid leukaemia. Lancet 2015, 385, 1447–1459. [Google Scholar] [CrossRef]
  119. Siveen, K.S.; Prabhu, K.S.; Achkar, I.W.; Kuttikrishnan, S.; Shyam, S.; Khan, A.Q.; Merhi, M.; Dermime, S.; Uddin, S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Mol. Cancer 2018, 17, 31. [Google Scholar] [CrossRef] [Green Version]
  120. Steegmann, J.L.; Baccarani, M.; Breccia, M.; Casado, L.F.; García-Gutiérrez, V.; Hochhaus, A.; Kim, D.W.; Kim, T.D.; Khoury, H.J.; Le Coutre, P.; et al. European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia 2016, 30, 1648–1671. [Google Scholar] [CrossRef] [Green Version]
  121. Westerweel, P.E.; Te Boekhorst, P.A.W.; Levin, M.D.; Cornelissen, J.J. New Approaches and Treatment Combinations for the Management of Chronic Myeloid Leukemia. Front. Oncol. 2019, 9, 665. [Google Scholar] [CrossRef]
  122. Kipps, T.J.; Stevenson, F.K.; Wu, C.J.; Croce, C.M.; Packham, G.; Wierda, W.G.; O’Brien, S.; Gribben, J.; Rai, K. Chronic lymphocytic leukaemia. Nat. Rev. Dis. Prim. 2017, 3, 16096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Hamblin, T.J.; Davis, Z.; Gardiner, A.; Oscier, D.G.; Stevenson, F.K. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 1999, 94, 1848–1854. [Google Scholar] [CrossRef] [PubMed]
  124. Rodríguez-Vicente, A.E.; Díaz, M.G.; Hernández-Rivas, J.M. Chronic lymphocytic leukemia: A clinical and molecular heterogenous disease. Cancer Genet. 2013, 206, 49–62. [Google Scholar] [CrossRef] [PubMed]
  125. Puiggros, A.; Blanco, G. Genetic abnormalities in chronic lymphocytic leukemia: Where we are and where we go. Biomed Res. Int. 2014, 2014, 435983. [Google Scholar] [CrossRef] [PubMed]
  126. Kiefer, Y.; Schulte, C.; Tiemann, M.; Bullerdiek, J. Chronic lymphocytic leukemia-associated chromosomal abnormalities and miRNA deregulation. Appl. Clin. Genet. 2012, 5, 21–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Nabhan, C.; Raca, G.; Wang, Y.L. Predicting Prognosis in Chronic Lymphocytic Leukemia in the Contemporary Era. Jama Oncol. 2015, 1, 965–974. [Google Scholar] [CrossRef]
  128. Kipps, T.J. Chronic lymphocytic leukemia. Curr. Opini. Hematol. 2000, 7, 223–234. [Google Scholar] [CrossRef] [Green Version]
  129. Döhner, H.; Stilgenbauer, S.; Benner, A.; Leupolt, E.; Kröber, A.; Bullinger, L.; Döhner, K.; Bentz, M.; Lichter, P. Genomic aberrations and survival in chronic lymphocytic leukemia. N. Engl. J. Med. 2000, 343, 1910–1916. [Google Scholar] [CrossRef] [Green Version]
  130. Haferlach, C.; Dicker, F.; Schnittger, S.; Kern, W.; Haferlach, T. Comprehensive genetic characterization of CLL: A study on 506 cases analysed with chromosome banding analysis, interphase FISH, IgV(H) status and immunophenotyping. Leukemia 2007, 21, 2442–2451. [Google Scholar] [CrossRef] [Green Version]
  131. Zent, C.S.; Burack, W.R. Mutations in chronic lymphocytic leukemia and how they affect therapy choice: Focus on NOTCH1, SF3B1, and TP53. Hematol. Am. Soc. Hematol. Educ. Program. 2014, 2014, 119–124. [Google Scholar] [CrossRef] [Green Version]
  132. Kipps, T.J.; Choi, M.Y. Targeted Therapy in Chronic Lymphocytic Leukemia. Cancer J. 2019, 25, 378–385. [Google Scholar] [CrossRef] [PubMed]
  133. Burger, J.A. Treatment of Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2020, 383, 460–473. [Google Scholar] [CrossRef]
  134. Doulatov, S.; Notta, F.; Laurenti, E.; Dick, J.E. Hematopoiesis: A human perspective. Cell Stem Cell 2012, 10, 120–136. [Google Scholar] [CrossRef] [Green Version]
  135. Vedi, A.; Santoro, A.; Dunant, C.F.; Dick, J.E.; Laurenti, E. Molecular landscapes of human hematopoietic stem cells in health and leukemia. Ann. N. Y. Acad. Sci. 2016, 1370, 5–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Signer, R.A.; Magee, J.A.; Salic, A.; Morrison, S.J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 2014, 509, 49–54. [Google Scholar] [CrossRef] [PubMed]
  137. Khateb, A.; Ronai, Z.A. Unfolded Protein Response in Leukemia: From Basic Understanding to Therapeutic Opportunities. Trends Cancer 2020, 6, 960–973. [Google Scholar] [CrossRef] [PubMed]
  138. Hidalgo San Jose, L.; Sunshine, M.J.; Dillingham, C.H.; Chua, B.A.; Kruta, M.; Hong, Y.; Hatters, D.M.; Signer, R.A.J. Modest Declines in Proteome Quality Impair Hematopoietic Stem Cell Self-Renewal. Cell Rep. 2020, 30, 69–80.e66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. van Galen, P.; Kreso, A.; Mbong, N.; Kent, D.G.; Fitzmaurice, T.; Chambers, J.E.; Xie, S.; Laurenti, E.; Hermans, K.; Eppert, K.; et al. The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress. Nature 2014, 510, 268–272. [Google Scholar] [CrossRef]
  140. van Galen, P.; Mbong, N.; Kreso, A.; Schoof, E.M.; Wagenblast, E.; Ng, S.W.K.; Krivdova, G.; Jin, L.; Nakauchi, H.; Dick, J.E. Integrated Stress Response Activity Marks Stem Cells in Normal Hematopoiesis and Leukemia. Cell Rep. 2018, 25, 1109–1117.e1105. [Google Scholar] [CrossRef] [Green Version]
  141. Miharada, K.; Sigurdsson, V.; Karlsson, S. Dppa5 improves hematopoietic stem cell activity by reducing endoplasmic reticulum stress. Cell Rep. 2014, 7, 1381–1392. [Google Scholar] [CrossRef] [Green Version]
  142. Chapple, R.H.; Hu, T.; Tseng, Y.J.; Liu, L.; Kitano, A.; Luu, V.; Hoegenauer, K.A.; Iwawaki, T.; Li, Q.; Nakada, D. ERalpha promotes murine hematopoietic regeneration through the Ire1alpha-mediated unfolded protein response. eLife 2018, 7, e31159. [Google Scholar] [CrossRef] [PubMed]
  143. Liu, L.; Zhao, M.; Jin, X.; Ney, G.; Yang, K.B.; Peng, F.; Cao, J.; Iwawaki, T.; Del Valle, J.; Chen, X.; et al. Adaptive endoplasmic reticulum stress signalling via IRE1alpha-XBP1 preserves self-renewal of haematopoietic and pre-leukaemic stem cells. Nat. Cell Biol. 2019, 21, 328–337. [Google Scholar] [CrossRef] [PubMed]
  144. Xie, S.Z.; Garcia-Prat, L.; Voisin, V.; Ferrari, R.; Gan, O.I.; Wagenblast, E.; Kaufmann, K.B.; Zeng, A.G.X.; Takayanagi, S.I.; Patel, I.; et al. Sphingolipid Modulation Activates Proteostasis Programs to Govern Human Hematopoietic Stem Cell Self-Renewal. Cell Stem Cell 2019, 25, 639–653.e637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Xu, L.; Liu, X.; Peng, F.; Zhang, W.; Zheng, L.; Ding, Y.; Gu, T.; Lv, K.; Wang, J.; Ortinau, L.; et al. Protein quality control through endoplasmic reticulum-associated degradation maintains haematopoietic stem cell identity and niche interactions. Nat. Cell Biol. 2020, 22, 1162–1169. [Google Scholar] [CrossRef]
  146. Rouault-Pierre, K.; Hamilton, A.; Bonnet, D. Effect of hypoxia-inducible factors in normal and leukemic stem cell regulation and their potential therapeutic impact. Expert Opin. Biol. Ther. 2016, 16, 463–476. [Google Scholar] [CrossRef]
  147. Sigurdsson, V.; Miharada, K. Regulation of unfolded protein response in hematopoietic stem cells. Int. J. Hematol. 2018, 107, 627–633. [Google Scholar] [CrossRef] [Green Version]
  148. Doultsinos, D.; Avril, T.; Lhomond, S.; Dejeans, N.; Guédat, P.; Chevet, E. Control of the Unfolded Protein Response in Health and Disease. Slas Discov. Adv. Life Sci. R D 2017, 22, 787–800. [Google Scholar] [CrossRef] [Green Version]
  149. Siwecka, N.; Rozpędek, W.; Pytel, D.; Wawrzynkiewicz, A.; Dziki, A.; Dziki, Ł.; Diehl, J.A.; Majsterek, I. Dual role of Endoplasmic Reticulum Stress-Mediated Unfolded Protein Response Signaling Pathway in Carcinogenesis. Int. J. Mol. Sci. 2019, 20, 4354. [Google Scholar] [CrossRef] [Green Version]
  150. Martelli, A.M.; Paganelli, F.; Chiarini, F.; Evangelisti, C. The Unfolded Protein Response: A Novel Therapeutic Target in Acute Leukemias. Cancers 2020, 12, 333. [Google Scholar] [CrossRef] [Green Version]
  151. Kharabi Masouleh, B.; Chevet, E.; Panse, J.; Jost, E.; O’Dwyer, M.; Bruemmendorf, T.H.; Samali, A. Drugging the unfolded protein response in acute leukemias. J. Hematol. Oncol. 2015, 8, 87. [Google Scholar] [CrossRef] [Green Version]
  152. Schardt, J.A.; Weber, D.; Eyholzer, M.; Mueller, B.U.; Pabst, T. Activation of the unfolded protein response is associated with favorable prognosis in acute myeloid leukemia. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 3834–3841. [Google Scholar] [CrossRef] [Green Version]
  153. Tanimura, A.; Yujiri, T.; Tanaka, Y.; Tanaka, M.; Mitani, N.; Nakamura, Y.; Ariyoshi, K.; Tanizawa, Y. Activation of the unfolded protein response in primary acute myeloid leukemia cells. Int. J. Hematol. 2011, 94, 300–302. [Google Scholar] [CrossRef] [PubMed]
  154. Krysov, S.; Steele, A.J.; Coelho, V.; Linley, A.; Sanchez Hidalgo, M.; Carter, M.; Potter, K.N.; Kennedy, B.; Duncombe, A.S.; Ashton-Key, M.; et al. Stimulation of surface IgM of chronic lymphocytic leukemia cells induces an unfolded protein response dependent on BTK and SYK. Blood 2014, 124, 3101–3109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kharabi, M.B.; Geng, H.; Hurtz, C.; Chan, L.N.; Logan, A.C.; Chang, M.S.; Huang, C.; Swaminathan, S.; Sun, H.; Paietta, E.; et al. Mechanistic rationale for targeting the unfolded protein response in pre-B acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. USA 2014, 111, E2219–E2228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Zhou, C.; Martinez, E.; Di Marcantonio, D.; Solanki-Patel, N.; Aghayev, T.; Peri, S.; Ferraro, F.; Skorski, T.; Scholl, C.; Fröhling, S.; et al. JUN is a key transcriptional regulator of the unfolded protein response in acute myeloid leukemia. Leukemia 2017, 31, 1196–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Sun, H.; Lin, D.C.; Guo, X.; Kharabi, M.B.; Gery, S.; Cao, Q.; Alkan, S.; Ikezoe, T.; Akiba, C.; Paquette, R.; et al. Inhibition of IRE1α-driven pro-survival pathways is a promising therapeutic application in acute myeloid leukemia. Oncotarget 2016, 7, 18736–18749. [Google Scholar] [CrossRef] [PubMed]
  158. Bujisic, B.; De Gassart, A. Impairment of both IRE1 expression and XBP1 activation is a hallmark of GCB DLBCL and contributes to tumor growth. Blood 2017, 129, 2420–2428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Hart, L.S.; Cunningham, J.T.; Datta, T.; Dey, S.; Tameire, F.; Lehman, S.L.; Qiu, B.; Zhang, H.; Cerniglia, G.; Bi, M.; et al. ER stress-mediated autophagy promotes Myc-dependent transformation and tumor growth. J. Clin. Investig. 2012, 122, 4621–4634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Brancolini, C.; Iuliano, L. Proteotoxic Stress and Cell Death in Cancer Cells. Cancers 2020, 12, 2385. [Google Scholar] [CrossRef]
  161. Uckun, F.M.; Qazi, S.; Ozer, Z.; Garner, A.L.; Pitt, J.; Ma, H.; Janda, K.D. Inducing apoptosis in chemotherapy-resistant B-lineage acute lymphoblastic leukaemia cells by targeting HSPA5, a master regulator of the anti-apoptotic unfolded protein response signalling network. Br. J. Haematol. 2011, 153, 741–752. [Google Scholar] [CrossRef]
  162. Kusio-Kobialka, M.; Podszywalow-Bartnicka, P.; Peidis, P.; Glodkowska-Mrowka, E.; Wolanin, K.; Leszak, G.; Seferynska, I.; Stoklosa, T.; Koromilas, A.E.; Piwocka, K. The PERK-eIF2α phosphorylation arm is a pro-survival pathway of BCR-ABL signaling and confers resistance to imatinib treatment in chronic myeloid leukemia cells. Cell Cycle 2012, 11, 4069–4078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Higa, A.; Taouji, S.; Lhomond, S.; Jensen, D.; Fernandez-Zapico, M.E.; Simpson, J.C.; Pasquet, J.M.; Schekman, R.; Chevet, E. Endoplasmic reticulum stress-activated transcription factor ATF6α requires the disulfide isomerase PDIA5 to modulate chemoresistance. Mol. Cell. Biol. 2014, 34, 1839–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Tang, C.H.; Ranatunga, S.; Kriss, C.L.; Cubitt, C.L.; Tao, J.; Pinilla-Ibarz, J.A.; Del Valle, J.R.; Hu, C.C. Inhibition of ER stress-associated IRE-1/XBP-1 pathway reduces leukemic cell survival. J. Clin. Investig. 2014, 124, 2585–2598. [Google Scholar] [CrossRef] [PubMed]
  165. Tanimura, A.; Yujiri, T.; Tanaka, Y.; Hatanaka, M.; Mitani, N.; Nakamura, Y.; Mori, K.; Tanizawa, Y. The anti-apoptotic role of the unfolded protein response in Bcr-Abl-positive leukemia cells. Leuk. Res. 2009, 33, 924–928. [Google Scholar] [CrossRef] [PubMed]
  166. Leclerc, G.M.; Zheng, S.; Leclerc, G.J.; DeSalvo, J.; Swords, R.T.; Barredo, J.C. The NEDD8-activating enzyme inhibitor pevonedistat activates the eIF2α and mTOR pathways inducing UPR-mediated cell death in acute lymphoblastic leukemia. Leuk. Res. 2016, 50, 1–10. [Google Scholar] [CrossRef]
  167. Huiting, L.N.; Samaha, Y.; Zhang, G.L.; Roderick, J.E.; Li, B.; Anderson, N.M.; Wang, Y.W.; Wang, L.; Laroche, F.; Choi, J.W.; et al. UFD1 contributes to MYC-mediated leukemia aggressiveness through suppression of the proapoptotic unfolded protein response. Leukemia 2018, 32, 2339–2351. [Google Scholar] [CrossRef]
  168. Wilhelm, T.; Bick, F.; Peters, K.; Mohta, V.; Tirosh, B.; Patterson, J.B.; Kharabi-Masouleh, B.; Huber, M. Infliction of proteotoxic stresses by impairment of the unfolded protein response or proteasomal inhibition as a therapeutic strategy for mast cell leukemia. Oncotarget 2018, 9, 2984–3000. [Google Scholar] [CrossRef] [Green Version]
  169. Vieri, M.; Preisinger, C.; Schemionek, M.; Salimi, A.; Patterson, J.B.; Samali, A.; Brummendorf, T.H.; Appelmann, I.; Kharabi Masouleh, B. Targeting of BCR-ABL1 and IRE1α induces synthetic lethality in Philadelphia-positive acute lymphoblastic leukemia. Carcinogenesis 2020, bgaa095. [Google Scholar] [CrossRef]
  170. Jang, J.E.; Eom, J.I.; Jeung, H.K.; Chung, H.; Kim, Y.R.; Kim, J.S.; Cheong, J.W.; Min, Y.H. PERK/NRF2 and autophagy form a resistance mechanism against G9a inhibition in leukemia stem cells. J. Exp. Clin. Cancer Res. CR 2020, 39, 66. [Google Scholar] [CrossRef] [Green Version]
  171. Nguyen, T.K.; Grant, S. Dinaciclib (SCH727965) inhibits the unfolded protein response through a CDK1- and 5-dependent mechanism. Mol. Cancer Ther. 2014, 13, 662–674. [Google Scholar] [CrossRef] [Green Version]
  172. Evangelisti, C.; Evangelisti, C.; Teti, G.; Chiarini, F.; Falconi, M.; Melchionda, F.; Pession, A.; Bertaina, A.; Locatelli, F.; McCubrey, J.A.; et al. Assessment of the effect of sphingosine kinase inhibitors on apoptosis, unfolded protein response and autophagy of T-cell acute lymphoblastic leukemia cells; indications for novel therapeutics. Oncotarget 2014, 5, 7886–7901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Rahmani, M.; Mayo, M.; Dash, R.; Sokhi, U.K.; Dmitriev, I.P.; Sarkar, D.; Dent, P.; Curiel, D.T.; Fisher, P.B.; Grant, S. Melanoma differentiation associated gene-7/interleukin-24 potently induces apoptosis in human myeloid leukemia cells through a process regulated by endoplasmic reticulum stress. Mol. Pharm. 2010, 78, 1096–1104. [Google Scholar] [CrossRef] [PubMed]
  174. Rahmani, M.; Davis, E.M.; Crabtree, T.R.; Habibi, J.R.; Nguyen, T.K.; Dent, P.; Grant, S. The kinase inhibitor sorafenib induces cell death through a process involving induction of endoplasmic reticulum stress. Mol. Cell. Biol. 2007, 27, 5499–5513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Zhang, X.H.; Wang, X.Y.; Zhou, Z.W.; Bai, H.; Shi, L.; Yang, Y.X.; Zhou, S.F.; Zhang, X.C. The combination of digoxin and GSK2606414 exerts synergistic anticancer activity against leukemia in vitro and in vivo. Biofactors 2017, 43, 812–820. [Google Scholar] [CrossRef] [PubMed]
  176. Trivedi, R.; Müller, G.A.; Rathore, M.S.; Mishra, D.P.; Dihazi, H. Anti-Leukemic Activity of Shikonin: Role of ERP57 in Shikonin Induced Apoptosis in Acute Myeloid Leukemia. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharm. 2016, 39, 604–616. [Google Scholar] [CrossRef] [PubMed]
  177. Zhou, J.; Bi, C.; Cheong, L.L.; Mahara, S.; Liu, S.C.; Tay, K.G.; Koh, T.L.; Yu, Q.; Chng, W.J. The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood 2011, 118, 2830–2839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Jiang, Y.; Zhang, Y.; Wark, L.; Ortiz, E.; Lim, S.; He, H.; Wang, W.; Medeiros, D.; Lin, D. Wolfberry Water Soluble Phytochemicals Down-Regulate ER Stress Biomarkers and Modulate Multiple Signaling Pathways Leading to Inhibition of Proliferation and Induction of Apoptosis in Jurkat Cells. J. Nutr. Food Sci. 2011, S2. [Google Scholar] [CrossRef] [Green Version]
  179. Leclerc, G.M.; Leclerc, G.J.; Kuznetsov, J.N.; DeSalvo, J.; Barredo, J.C. Metformin induces apoptosis through AMPK-dependent inhibition of UPR signaling in ALL lymphoblasts. PLoS ONE 2013, 8, e74420. [Google Scholar] [CrossRef] [PubMed]
  180. Gu, L.; Yi, Z.; Zhang, Y.; Ma, Z.; Zhu, Y.; Gao, J. Low dose of 2-deoxy-D-glucose kills acute lymphoblastic leukemia cells and reverses glucocorticoid resistance via N-linked glycosylation inhibition under normoxia. Oncotarget 2017, 8, 30978–30991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. DeSalvo, J.; Kuznetsov, J.N.; Du, J.; Leclerc, G.M.; Leclerc, G.J.; Lampidis, T.J.; Barredo, J.C. Inhibition of Akt potentiates 2-DG-induced apoptosis via downregulation of UPR in acute lymphoblastic leukemia. Mol. Cancer Res. MCR 2012, 10, 969–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Jiang, Q.; Li, F.; Shi, K.; Wu, P.; An, J.; Yang, Y.; Xu, C. Involvement of p38 in signal switching from autophagy to apoptosis via the PERK/eIF2α/ATF4 axis in selenite-treated NB4 cells. Cell Death Dis. 2014, 5, e1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Rong, C.; Wei, W.; Yu-Hong, T. Asperuloside exhibits a novel anti-leukemic activity by triggering ER stress-regulated apoptosis via targeting GRP78. Biomed. Pharm. Biomed. Pharm. 2020, 125, 109819. [Google Scholar] [CrossRef] [PubMed]
  184. Maioral, M.F.; Stefanes, N.M.; Neuenfeldt, P.D.; Chiaradia-Delatorre, L.D.; Nunes, R.J.; Santos-Silva, M.C. Aldehyde biphenyl chalcones induce immunogenic apoptotic-like cell death and are promising new safe compounds against a wide range of hematologic cancers. Future Med. Chem. 2020, 12, 673–688. [Google Scholar] [CrossRef] [PubMed]
  185. Fang, W.; Xin, W.; Na, L.; Juan, L.; Lin, Z.; Lingyun, H.; Ai, F.; Zhonglin, W.; Yawen, W. Prolonged unfolded protein reaction is involved in the induction of CML cell death upon oprozomib treatment. Cancer Sci. 2020, 112, 133–143. [Google Scholar] [CrossRef]
  186. El Dor, M.; Dakik, H.; Polomski, M.; Haudebourg, E.; Brachet, M.; Gouilleux, F.; Prié, G.; Zibara, K.; Mazurier, F. VAS3947 Induces UPR-Mediated Apoptosis through Cysteine Thiol Alkylation in AML Cell Lines. Int. J. Mol. Sci. 2020, 21, 5470. [Google Scholar] [CrossRef]
  187. Hu, X.; Cai, J.; Zhu, J.; Lang, W.; Zhong, J.; Zhong, H.; Chen, F. Arsenic trioxide potentiates Gilteritinib-induced apoptosis in FLT3-ITD positive leukemic cells via IRE1a-JNK-mediated endoplasmic reticulum stress. Cancer Cell Int. 2020, 20, 250. [Google Scholar] [CrossRef]
  188. Chu, X.; Zhong, L.; Yu, L.; Xiong, L.; Li, J.; Dan, W.; Ye, J.; Liu, C.; Luo, X.; Liu, B. GSK-J4 induces cell cycle arrest and apoptosis via ER stress and the synergism between GSK-J4 and decitabine in acute myeloid leukemia KG-1a cells. Cancer Cell Int. 2020, 20, 209. [Google Scholar] [CrossRef]
  189. Bian, T.; Tagmount, A.; Vulpe, C.; Vijendra, K.C.; Xing, C. CXL146, a Novel 4H-Chromene Derivative, Targets GRP78 to Selectively Eliminate Multidrug-Resistant Cancer Cells. Mol. Pharm. 2020, 97, 402–408. [Google Scholar] [CrossRef] [Green Version]
  190. Matsumoto, T.; Jimi, S.; Migita, K.; Terada, K.; Mori, M.; Takamatsu, Y.; Suzumiya, J.; Hara, S. FF-10501 induces caspase-8-mediated apoptotic and endoplasmic reticulum stress-mediated necrotic cell death in hematological malignant cells. Int. J. Hematol. 2019, 110, 606–617. [Google Scholar] [CrossRef]
  191. Masciarelli, S.; Capuano, E.; Ottone, T.; Divona, M.; Lavorgna, S.; Liccardo, F.; Śniegocka, M.; Travaglini, S.; Noguera, N.I.; Picardi, A.; et al. Retinoic acid synergizes with the unfolded protein response and oxidative stress to induce cell death in FLT3-ITD+ AML. Blood Adv. 2019, 3, 4155–4160. [Google Scholar] [CrossRef] [Green Version]
  192. Masciarelli, S.; Capuano, E.; Ottone, T.; Divona, M.; De Panfilis, S.; Banella, C.; Noguera, N.I.; Picardi, A.; Fontemaggi, G.; Blandino, G.; et al. Retinoic acid and arsenic trioxide sensitize acute promyelocytic leukemia cells to ER stress. Leukemia 2018, 32, 285–294. [Google Scholar] [CrossRef] [Green Version]
  193. Mallick, D.J.; Soderquist, R.S.; Bates, D.; Eastman, A. Confounding off-target effects of BH3 mimetics at commonly used concentrations: MIM1, UMI-77, and A-1210477. Cell Death Dis. 2019, 10, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Hsiao, Y.C.; Peng, S.F.; Lai, K.C.; Liao, C.L.; Huang, Y.P.; Lin, C.C.; Lin, M.L.; Liu, K.C.; Tsai, C.C.; Ma, Y.S.; et al. Genistein induces apoptosis in vitro and has antitumor activity against human leukemia HL-60 cancer cell xenograft growth in vivo. Environ. Toxicol. 2019, 34, 443–456. [Google Scholar] [CrossRef] [PubMed]
  195. Yang, Y.; Wang, G.; Wu, W.; Yao, S.; Han, X.; He, D.; He, J.; Zheng, G.; Zhao, Y.; Cai, Z.; et al. Camalexin Induces Apoptosis via the ROS-ER Stress-Mitochondrial Apoptosis Pathway in AML Cells. Oxidative Med. Cell. Longev. 2018, 2018, 7426950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Li, Z.; Wu, J.; Sheng, L. Ibrutinib improves the development of acute lymphoblastic leukemia by activating endoplasmic reticulum stress-induced cell death. Pharmazie 2018, 73, 294–299. [Google Scholar] [CrossRef]
  197. Mazumder, A.; Lee, J.Y.; Talhi, O.; Cerella, C.; Chateauvieux, S.; Gaigneaux, A.; Hong, C.R.; Kang, H.J.; Lee, Y.; Kim, K.W.; et al. Hydroxycoumarin OT-55 kills CML cells alone or in synergy with imatinib or Synribo: Involvement of ER stress and DAMP release. Cancer Lett. 2018, 438, 197–218. [Google Scholar] [CrossRef]
  198. Florean, C.; Kim, K.R.; Schnekenburger, M.; Kim, H.J.; Moriou, C.; Debitus, C.; Dicato, M.; Al-Mourabit, A.; Han, B.W.; Diederich, M. Synergistic AML Cell Death Induction by Marine Cytotoxin (+)-1(R), 6(S), 1′(R), 6′(S), 11(R), 17(S)-Fistularin-3 and Bcl-2 Inhibitor Venetoclax. Mar. Drugs 2018, 16, 518. [Google Scholar] [CrossRef] [Green Version]
  199. Wang, X.Y.; Zhang, X.H.; Peng, L.; Liu, Z.; Yang, Y.X.; He, Z.X.; Dang, H.W.; Zhou, S.F. Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. Am. J. Trans. Res. 2017, 9, 4652–4672. [Google Scholar]
  200. Mitsuhashi, Y.; Furusawa, Y.; Aradate, T.; Zhao, Q.L.; Moniruzzaman, R.; Kanamori, M.; Noguchi, K.; Kondo, T. 3-O-trans-p-coumaroyl-alphitolic acid, a triterpenoid from Zizyphus jujuba, leads to apoptotic cell death in human leukemia cells through reactive oxygen species production and activation of the unfolded protein response. PLoS ONE 2017, 12, e0183712. [Google Scholar] [CrossRef] [Green Version]
  201. Meier-Stephenson, V.; Riemer, J.; Narendran, A. The HIV protease inhibitor, nelfinavir, as a novel therapeutic approach for the treatment of refractory pediatric leukemia. Oncotargets 2017, 10, 2581–2593. [Google Scholar] [CrossRef] [Green Version]
  202. Mahoney, E.; Maddocks, K.; Flynn, J.; Jones, J.; Cole, S.L.; Zhang, X.; Byrd, J.C.; Johnson, A.J. Identification of endoplasmic reticulum stress-inducing agents by antagonizing autophagy: A new potential strategy for identification of anti-cancer therapeutics in B-cell malignancies. Leuk. Lymphoma 2013, 54, 2685–2692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Gugliotta, G.; Sudo, M.; Cao, Q.; Lin, D.C.; Sun, H.; Takao, S.; Le Moigne, R.; Rolfe, M.; Gery, S.; Müschen, M.; et al. Valosin-Containing Protein/p97 as a Novel Therapeutic Target in Acute Lymphoblastic Leukemia. Neoplasia 2017, 19, 750–761. [Google Scholar] [CrossRef] [PubMed]
  204. Tsitsipatis, D.; Jayavelu, A.K.; Müller, J.P.; Bauer, R.; Schmidt-Arras, D.; Mahboobi, S.; Schnöder, T.M.; Heidel, F.; Böhmer, F.D. Synergistic killing of FLT3ITD-positive AML cells by combined inhibition of tyrosine-kinase activity and N-glycosylation. Oncotarget 2017, 8, 26613–26624. [Google Scholar] [CrossRef] [PubMed]
  205. Wu, C.F.; Seo, E.J.; Klauck, S.M.; Efferth, T. Cryptotanshinone deregulates unfolded protein response and eukaryotic initiation factor signaling in acute lymphoblastic leukemia cells. Phytomed. Int. J. Phytother. Phytopharm. 2016, 23, 174–180. [Google Scholar] [CrossRef] [PubMed]
  206. Wang, J.; Wang, Q.L.; Nong, X.H.; Zhang, X.Y.; Xu, X.Y.; Qi, S.H.; Wang, Y.F. Oxalicumone A, a new dihydrothiophene-condensed sulfur chromone induces apoptosis in leukemia cells through endoplasmic reticulum stress pathway. Eur. J. Pharm. 2016, 783, 47–55. [Google Scholar] [CrossRef]
  207. Lamothe, B.; Wierda, W.G.; Keating, M.J.; Gandhi, V. Carfilzomib Triggers Cell Death in Chronic Lymphocytic Leukemia by Inducing Proapoptotic and Endoplasmic Reticulum Stress Responses. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 4712–4726. [Google Scholar] [CrossRef] [Green Version]
  208. Zhou, L.; Jiang, L.; Xu, M.; Liu, Q.; Gao, N.; Li, P.; Liu, E.H. Miltirone exhibits antileukemic activity by ROS-mediated endoplasmic reticulum stress and mitochondrial dysfunction pathways. Sci. Rep. 2016, 6, 20585. [Google Scholar] [CrossRef] [Green Version]
  209. Wang, F.F.; Liu, M.Z.; Sui, Y.; Cao, Q.; Yan, B.; Jin, M.L.; Mo, X. Deficiency of SUMO-specific protease 1 induces arsenic trioxide-mediated apoptosis by regulating XBP1 activity in human acute promyelocytic leukemia. Oncol. Lett. 2016, 12, 3755–3762. [Google Scholar] [CrossRef] [Green Version]
  210. Chen, J.; Wei, H.; Xie, B.; Wang, B.; Cheng, J.; Cheng, J. Endoplasmic reticulum stress contributes to arsenic trioxide-induced apoptosis in drug-sensitive and -resistant leukemia cells. Leuk. Res. 2012, 36, 1526–1535. [Google Scholar] [CrossRef]
  211. Rosilio, C.; Nebout, M.; Imbert, V.; Griessinger, E.; Neffati, Z.; Benadiba, J.; Hagenbeek, T.; Spits, H.; Reverso, J.; Ambrosetti, D.; et al. L-type amino-acid transporter 1 (LAT1): A therapeutic target supporting growth and survival of T-cell lymphoblastic lymphoma/T-cell acute lymphoblastic leukemia. Leukemia 2015, 29, 1253–1266. [Google Scholar] [CrossRef]
  212. Hu, C.; Xu, M.; Qin, R.; Chen, W.; Xu, X. Wogonin induces apoptosis and endoplasmic reticulum stress in HL-60 leukemia cells through inhibition of the PI3K-AKT signaling pathway. Oncol. Rep. 2015, 33, 3146–3154. [Google Scholar] [CrossRef] [PubMed]
  213. Joo, J.H.; Ueda, E.; Bortner, C.D.; Yang, X.P.; Liao, G.; Jetten, A.M. Farnesol activates the intrinsic pathway of apoptosis and the ATF4-ATF3-CHOP cascade of ER stress in human T lymphoblastic leukemia Molt4 cells. Biochem. Pharm. 2015, 97, 256–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Cagnetta, A.; Caffa, I.; Acharya, C.; Soncini, D.; Acharya, P.; Adamia, S.; Pierri, I.; Bergamaschi, M.; Garuti, A.; Fraternali, G.; et al. APO866 Increases Antitumor Activity of Cyclosporin-A by Inducing Mitochondrial and Endoplasmic Reticulum Stress in Leukemia Cells. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 3934–3945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Takenokuchi, M.; Miyamoto, K.; Saigo, K.; Taniguchi, T. Bortezomib Causes ER Stress-related Death of Acute Promyelocytic Leukemia Cells Through Excessive Accumulation of PML-RARA. Anticancer Res. 2015, 35, 3307–3316. [Google Scholar] [PubMed]
  216. Buontempo, F.; Orsini, E.; Martins, L.R.; Antunes, I.; Lonetti, A.; Chiarini, F.; Tabellini, G.; Evangelisti, C.; Evangelisti, C.; Melchionda, F.; et al. Cytotoxic activity of the casein kinase 2 inhibitor CX-4945 against T-cell acute lymphoblastic leukemia: Targeting the unfolded protein response signaling. Leukemia 2014, 28, 543–553. [Google Scholar] [CrossRef]
  217. Winter, E.; Dal Pizzol, C.; Filippin-Monteiro, F.B.; Brondani, P.; Silva, A.M.; Silva, A.H.; Bonacorso, H.G.; Martins, M.A.; Zanatta, N.; Creczynski-Pasa, T.B. Antitumoral activity of a trichloromethyl pyrimidine analogue: Molecular cross-talk between intrinsic and extrinsic apoptosis. Chem. Res. Toxicol. 2014, 27, 1040–1049. [Google Scholar] [CrossRef]
  218. Winter, E.; Chiaradia, L.D.; Silva, A.H.; Nunes, R.J.; Yunes, R.A.; Creczynski-Pasa, T.B. Involvement of extrinsic and intrinsic apoptotic pathways together with endoplasmic reticulum stress in cell death induced by naphthylchalcones in a leukemic cell line: Advantages of multi-target action. Toxicol. Vitr. 2014, 28, 769–777. [Google Scholar] [CrossRef] [Green Version]
  219. Soderquist, R.; Pletnev, A.A.; Danilov, A.V.; Eastman, A. The putative BH3 mimetic S1 sensitizes leukemia to ABT-737 by increasing reactive oxygen species, inducing endoplasmic reticulum stress, and upregulating the BH3-only protein NOXA. Apoptosis Int. J. Program. Cell Death 2014, 19, 201–209. [Google Scholar] [CrossRef] [Green Version]
  220. Soderquist, R.S.; Danilov, A.V.; Eastman, A. Gossypol increases expression of the pro-apoptotic BH3-only protein NOXA through a novel mechanism involving phospholipase A2, cytoplasmic calcium, and endoplasmic reticulum stress. J. Biol. Chem. 2014, 289, 16190–16199. [Google Scholar] [CrossRef] [Green Version]
  221. Li, H.; Chang, G.; Wang, J.; Wang, L.; Jin, W.; Lin, Y.; Yan, Y.; Wang, R.; Gao, W.; Ma, L.; et al. Cariporide sensitizes leukemic cells to tumor necrosis factor related apoptosis-inducing ligand by up-regulation of death receptor 5 via endoplasmic reticulum stress-CCAAT/enhancer binding protein homologous protein dependent mechanism. Leuk. Lymphoma 2014, 55, 2135–2140. [Google Scholar] [CrossRef]
  222. Fiskus, W.; Saba, N.; Shen, M.; Ghias, M.; Liu, J.; Gupta, S.D.; Chauhan, L.; Rao, R.; Gunewardena, S.; Schorno, K.; et al. Auranofin induces lethal oxidative and endoplasmic reticulum stress and exerts potent preclinical activity against chronic lymphocytic leukemia. Cancer Res. 2014, 74, 2520–2532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Bortolozzi, R.; Viola, G.; Porcù, E.; Consolaro, F.; Marzano, C.; Pellei, M.; Gandin, V.; Basso, G. A novel copper(I) complex induces ER-stress-mediated apoptosis and sensitizes B-acute lymphoblastic leukemia cells to chemotherapeutic agents. Oncotarget 2014, 5, 5978–5991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Rosati, E.; Sabatini, R.; De Falco, F.; Del Papa, B.; Falzetti, F.; Di Ianni, M.; Cavalli, L.; Fettucciari, K.; Bartoli, A.; Screpanti, I.; et al. γ-Secretase inhibitor I induces apoptosis in chronic lymphocytic leukemia cells by proteasome inhibition, endoplasmic reticulum stress increase and notch down-regulation. Int. J. Cancer 2013, 132, 1940–1953. [Google Scholar] [CrossRef] [PubMed]
  225. Huang, A.C.; Chang, C.L.; Yu, C.S.; Chen, P.Y.; Yang, J.S.; Ji, B.C.; Lin, T.P.; Chiu, C.F.; Yeh, S.P.; Huang, Y.P.; et al. Induction of apoptosis by curcumin in murine myelomonocytic leukemia WEHI-3 cells is mediated via endoplasmic reticulum stress and mitochondria-dependent pathways. Environ. Toxicol. 2013, 28, 255–266. [Google Scholar] [CrossRef]
  226. Ng, A.P.; Chng, W.J.; Khan, M. Curcumin sensitizes acute promyelocytic leukemia cells to unfolded protein response-induced apoptosis by blocking the loss of misfolded N-CoR protein. Mol. Cancer Res. MCR 2011, 9, 878–888. [Google Scholar] [CrossRef] [Green Version]
  227. Pae, H.O.; Jeong, S.O.; Jeong, G.S.; Kim, K.M.; Kim, H.S.; Kim, S.A.; Kim, Y.C.; Kang, S.D.; Kim, B.N.; Chung, H.T. Curcumin induces pro-apoptotic endoplasmic reticulum stress in human leukemia HL-60 cells. Biochem. Biophys. Res. Commun. 2007, 353, 1040–1045. [Google Scholar] [CrossRef]
  228. de Sá Bacelar, T.; da Silva, A.J.; Costa, P.R.; Rumjanek, V.M. The pterocarpanquinone LQB 118 induces apoptosis in tumor cells through the intrinsic pathway and the endoplasmic reticulum stress pathway. Anti-Cancer Drugs 2013, 24, 73–83. [Google Scholar] [CrossRef]
  229. Mahoney, E.; Byrd, J.C.; Johnson, A.J. Autophagy and ER stress play an essential role in the mechanism of action and drug resistance of the cyclin-dependent kinase inhibitor flavopiridol. Autophagy 2013, 9, 434–435. [Google Scholar] [CrossRef]
  230. Yu, C.S.; Huang, A.C.; Yang, J.S.; Yu, C.C.; Lin, C.C.; Chung, H.K.; Huang, Y.P.; Chueh, F.S.; Chung, J.G. Safrole induces G0/G1 phase arrest via inhibition of cyclin E and provokes apoptosis through endoplasmic reticulum stress and mitochondrion-dependent pathways in human leukemia HL-60 cells. Anticancer Res. 2012, 32, 1671–1679. [Google Scholar]
  231. Penna, F.; Pin, F.; Costamagna, D.; Reffo, P.; Baccino, F.M.; Bonelli, G.; Costelli, P. Caspase 2 activation and ER stress drive rapid Jurkat cell apoptosis by clofibrate. PLoS ONE 2012, 7, e45327. [Google Scholar] [CrossRef]
  232. Park, Y.K.; Do, Y.R.; Jang, B.C. Apoptosis of K562 leukemia cells by Abnobaviscum F®, a European mistletoe extract. Oncol. Rep. 2012, 28, 2227–2232. [Google Scholar] [CrossRef] [PubMed]
  233. Lu, C.C.; Yang, J.S.; Chiang, J.H.; Hour, M.J.; Lin, K.L.; Lin, J.J.; Huang, W.W.; Tsuzuki, M.; Lee, T.H.; Chung, J.G. Novel quinazolinone MJ-29 triggers endoplasmic reticulum stress and intrinsic apoptosis in murine leukemia WEHI-3 cells and inhibits leukemic mice. PLoS ONE 2012, 7, e36831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Pattacini, L.; Mancini, M.; Mazzacurati, L.; Brusa, G.; Benvenuti, M.; Martinelli, G.; Baccarani, M.; Santucci, M.A. Endoplasmic reticulum stress initiates apoptotic death induced by STI571 inhibition of p210 bcr-abl tyrosine kinase. Leuk. Res. 2004, 28, 191–202. [Google Scholar] [CrossRef]
  235. Chueh, F.S.; Hsiao, Y.T.; Chang, S.J.; Wu, P.P.; Yang, J.S.; Lin, J.J.; Chung, J.G.; Lai, T.Y. Glycyrrhizic acid induces apoptosis in WEHI-3 mouse leukemia cells through the caspase- and mitochondria-dependent pathways. Oncol. Rep. 2012, 28, 2069–2076. [Google Scholar] [CrossRef] [Green Version]
  236. Lin, J.J.; Hsu, H.Y.; Yang, J.S.; Lu, K.W.; Wu, R.S.; Wu, K.C.; Lai, T.Y.; Chen, P.Y.; Ma, C.Y.; Wood, W.G.; et al. Molecular evidence of anti-leukemia activity of gypenosides on human myeloid leukemia HL-60 cells in vitro and in vivo using a HL-60 cells murine xenograft model. Phytomedicine Int. J. Phytother. Phytopharm. 2011, 18, 1075–1085. [Google Scholar] [CrossRef]
  237. Kuznetsov, J.N.; Leclerc, G.J.; Leclerc, G.M.; Barredo, J.C. AMPK and Akt determine apoptotic cell death following perturbations of one-carbon metabolism by regulating ER stress in acute lymphoblastic leukemia. Mol. Cancer Ther. 2011, 10, 437–447. [Google Scholar] [CrossRef] [Green Version]
  238. Chang, Y.C.; Lai, T.Y.; Yu, C.S.; Chen, H.Y.; Yang, J.S.; Chueh, F.S.; Lu, C.C.; Chiang, J.H.; Huang, W.W.; Ma, C.Y.; et al. Emodin Induces Apoptotic Death in Murine Myelomonocytic Leukemia WEHI-3 Cells In Vitro and Enhances Phagocytosis in Leukemia Mice In Vivo. Evid. Based Complementary Altern. Med. ECAM 2011, 2011, 523596. [Google Scholar] [CrossRef]
  239. Anshu, A.; Thomas, S.; Agarwal, P.; Ibarra-Rivera, T.R.; Pirrung, M.C.; Schönthal, A.H. Novel proteasome-inhibitory syrbactin analogs inducing endoplasmic reticulum stress and apoptosis in hematological tumor cell lines. Biochem. Pharmacol. 2011, 82, 600–609. [Google Scholar] [CrossRef]
  240. Albershardt, T.C.; Salerni, B.L.; Soderquist, R.S.; Bates, D.J.; Pletnev, A.A.; Kisselev, A.F.; Eastman, A. Multiple BH3 mimetics antagonize antiapoptotic MCL1 protein by inducing the endoplasmic reticulum stress response and up-regulating BH3-only protein NOXA. J. Biol. Chem. 2011, 286, 24882–24895. [Google Scholar] [CrossRef] [Green Version]
  241. Wagh, V.; Mishra, P.; Thakkar, A.; Shinde, V.; Sharma, S.; Padigaru, M.; Joshi, K. Antitumor activity of NPB001-05, an orally active inhibitor of Bcr-Abl tyrosine kinase. Front. Biosci. 2011, 3, 1349–1364. [Google Scholar] [CrossRef]
  242. Yaari-Stark, S.; Shaked, M.; Nevo-Caspi, Y.; Jacob-Hircsh, J.; Shamir, R.; Rechavi, G.; Kloog, Y. Ras inhibits endoplasmic reticulum stress in human cancer cells with amplified Myc. Int. J. Cancer 2010, 126, 2268–2281. [Google Scholar] [CrossRef] [PubMed]
  243. Xu, G.W.; Ali, M.; Wood, T.E.; Wong, D.; Maclean, N.; Wang, X.; Gronda, M.; Skrtic, M.; Li, X.; Hurren, R.; et al. The ubiquitin-activating enzyme E1 as a therapeutic target for the treatment of leukemia and multiple myeloma. Blood 2010, 115, 2251–2259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Popovic, S.; Baskic, D.; Djurdjevic, P.; Zelen, I.; Mitrovic, M.; Nikolic, I.; Avramovic, D.; Radenkovic, M.; Arsenijevic, N. Endoplasmic reticulum stress associated with caspases-4 and -2 mediates korbazol-induced B-chronic lymphocytic leukemia cell apoptosis. J. B.U. Off. J. Balk. Union Oncol. 2010, 15, 783–790. [Google Scholar]
  245. Lust, S.; Vanhoecke, B.; Van Gele, M.; Philippé, J.; Bracke, M.; Offner, F. The flavonoid tangeretin activates the unfolded protein response and synergizes with imatinib in the erythroleukemia cell line K562. Mol. Nutr. Food Res. 2010, 54, 823–832. [Google Scholar] [CrossRef] [PubMed]
  246. Lee, M.S.; Cherla, R.P.; Lentz, E.K.; Leyva-Illades, D.; Tesh, V.L. Signaling through C/EBP homologous protein and death receptor 5 and calpain activation differentially regulate THP-1 cell maturation-dependent apoptosis induced by Shiga toxin type 1. Infect. Immun. 2010, 78, 3378–3391. [Google Scholar] [CrossRef] [Green Version]
  247. Slagsvold, J.E.; Pettersen, C.H.; Follestad, T.; Krokan, H.E.; Schønberg, S.A. The antiproliferative effect of EPA in HL60 cells is mediated by alterations in calcium homeostasis. Lipids 2009, 44, 103–113. [Google Scholar] [CrossRef]
  248. Lust, S.; Vanhoecke, B.; Van Gelle, M.; Boelens, J.; van Melckebeke, H.; Kaileh, M.; Vanden Berghe, W.; Haegeman, G.; Philippé, J.; Bracke, M.; et al. Xanthohumol activates the proapoptotic arm of the unfolded protein response in chronic lymphocytic leukemia. Anticancer Res. 2009, 29, 3797–3805. [Google Scholar]
  249. Huang, W.C.; Lin, Y.S.; Chen, C.L.; Wang, C.Y.; Chiu, W.H.; Lin, C.F. Glycogen synthase kinase-3beta mediates endoplasmic reticulum stress-induced lysosomal apoptosis in leukemia. J. Pharmacol. Exp. Ther. 2009, 329, 524–531. [Google Scholar] [CrossRef] [Green Version]
  250. Zhang, Q.Y.; Mao, J.H.; Liu, P.; Huang, Q.H.; Lu, J.; Xie, Y.Y.; Weng, L.; Zhang, Y.; Chen, Q.; Chen, S.J.; et al. A systems biology understanding of the synergistic effects of arsenic sulfide and Imatinib in BCR/ABL-associated leukemia. Proc. Natl. Acad. Sci. USA 2009, 106, 3378–3383. [Google Scholar] [CrossRef] [Green Version]
  251. Wang, K.; Fang, H.; Xiao, D.; Zhu, X.; He, M.; Pan, X.; Shi, J.; Zhang, H.; Jia, X.; Du, Y.; et al. Converting redox signaling to apoptotic activities by stress-responsive regulators HSF1 and NRF2 in fenretinide treated cancer cells. PLoS ONE 2009, 4, e7538. [Google Scholar] [CrossRef] [Green Version]
  252. Lai, W.L.; Wong, N.S. The PERK/eIF2 alpha signaling pathway of Unfolded Protein Response is essential for N-(4-hydroxyphenyl)retinamide (4HPR)-induced cytotoxicity in cancer cells. Exp. Cell Res. 2008, 314, 1667–1682. [Google Scholar] [CrossRef] [PubMed]
  253. Townsend, D.M.; Manevich, Y.; He, L.; Xiong, Y.; Bowers, R.R., Jr.; Hutchens, S.; Tew, K.D. Nitrosative stress-induced s-glutathionylation of protein disulfide isomerase leads to activation of the unfolded protein response. Cancer Res. 2009, 69, 7626–7634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Dodo, K.; Minato, T.; Noguchi-Yachide, T.; Suganuma, M.; Hashimoto, Y. Antiproliferative and apoptosis-inducing activities of alkyl gallate and gallamide derivatives related to (-)-epigallocatechin gallate. Bioorganic Med. Chem. 2008, 16, 7975–7982. [Google Scholar] [CrossRef] [PubMed]
  255. Li, J.; Xia, X.; Ke, Y.; Nie, H.; Smith, M.A.; Zhu, X. Trichosanthin induced apoptosis in HL-60 cells via mitochondrial and endoplasmic reticulum stress signaling pathways. Biochim. Biophys. Acta 2007, 1770, 1169–1180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Jun, D.Y.; Kim, J.S.; Park, H.S.; Han, C.R.; Fang, Z.; Woo, M.H.; Rhee, I.K.; Kim, Y.H. Apoptogenic activity of auraptene of Zanthoxylum schinifolium toward human acute leukemia Jurkat T cells is associated with ER stress-mediated caspase-8 activation that stimulates mitochondria-dependent or -independent caspase cascade. Carcinogenesis 2007, 28, 1303–1313. [Google Scholar] [CrossRef] [PubMed]
  257. Anding, A.L.; Chapman, J.S.; Barnett, D.W.; Curley, R.W., Jr.; Clagett-Dame, M. The unhydrolyzable fenretinide analogue 4-hydroxybenzylretinone induces the proapoptotic genes GADD153 (CHOP) and Bcl-2-binding component 3 (PUMA) and apoptosis that is caspase- dependent and independent of the retinoic acid receptor. Cancer Res. 2007, 67, 6270–6277. [Google Scholar] [CrossRef] [Green Version]
  258. Yanamandra, N.; Colaco, N.M.; Parquet, N.A.; Buzzeo, R.W.; Boulware, D.; Wright, G.; Perez, L.E.; Dalton, W.S.; Beaupre, D.M. Tipifarnib and bortezomib are synergistic and overcome cell adhesion-mediated drug resistance in multiple myeloma and acute myeloid leukemia. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 591–599. [Google Scholar] [CrossRef] [Green Version]
  259. Ng, A.P.; Howe Fong, J.; Sijin Nin, D.; Hirpara, J.L.; Asou, N.; Chen, C.S.; Pervaiz, S.; Khan, M. Cleavage of misfolded nuclear receptor corepressor confers resistance to unfolded protein response-induced apoptosis. Cancer Res. 2006, 66, 9903–9912. [Google Scholar] [CrossRef] [Green Version]
  260. Feng, X.Q.; You, Y.; Xiao, J.; Zou, P. Thapsigargin-induced apoptosis of K562 cells and its mechanism. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2006, 14, 25–30. [Google Scholar]
  261. Feng, X.Q.; Xiao, J.; Nie, S.M.; Liu, F.; You, Y.; Zou, P. Expression and significance of melanoma antigen gene-3 in endoplasmic reticulum stress-induced apoptosis of K562 cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2005, 13, 741–745. [Google Scholar]
  262. Du, Y.; Wang, K.; Fang, H.; Li, J.; Xiao, D.; Zheng, P.; Chen, Y.; Fan, H.; Pan, X.; Zhao, C.; et al. Coordination of intrinsic, extrinsic, and endoplasmic reticulum-mediated apoptosis by imatinib mesylate combined with arsenic trioxide in chronic myeloid leukemia. Blood 2006, 107, 1582–1590. [Google Scholar] [CrossRef] [PubMed]
  263. Anether, G.; Tinhofer, I.; Senfter, M.; Greil, R. Tetrocarcin-A—Induced ER stress mediates apoptosis in B-CLL cells via a Bcl-2--independent pathway. Blood 2003, 101, 4561–4568. [Google Scholar] [CrossRef] [PubMed]
  264. Tinhofer, I.; Anether, G.; Senfter, M.; Pfaller, K.; Bernhard, D.; Hara, M.; Greil, R. Stressful death of T-ALL tumor cells after treatment with the anti-tumor agent Tetrocarcin-A. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2002, 16, 1295–1297. [Google Scholar] [CrossRef]
  265. Walczak, A.; Gradzik, K.; Kabzinski, J. The Role of the ER-Induced UPR Pathway and the Efficacy of Its Inhibitors and Inducers in the Inhibition of Tumor Progression. Oxidative Med. Cell. Longev. 2019, 2019, 5729710. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The different UPR effectors and their modes of action. In the basal state, the three UPR effector transmembrane proteins (PERK, ATF6, and IRE-1) are maintained inactive through their interaction with the protein chaperone BiP. The accumulation of misfolded proteins in the ER lumen results in dissociation of BiP and activation of UPR. (1) PERK dimerizes and phosphorylates the eIF2α subunit, leading to a global inhibition of translation initiation. Specific mRNA subsets, containing cis-acting elements in their 5′UTR, such as uORF and IRES, escape translational inhibition triggered by eIF2 phosphorylation. (2) IRE-1 initiates an unconventional splicing of XBP-1 mRNA. IRE1α cleaves Xbp1u mRNA within two stem-loop structures, leading to excision of 26 nucleotides. Subsequent ligation of the Xbp1 mRNA by the tRNA ligase RTCB results in a frame shift and allows the translation of the active transcription factor XBP1s, which is imported into the nucleus and activates the expression of target genes. IRE1α mediates also the degradation of some RNAs (this mechanism has been called RIDD for Regulated Ire1-Dependent Decay). (3) BIP dissociation from ATF6 exposes its Golgi Localization Signal. ATF6 is translocated to the Golgi apparatus where proteolysis releases its transcription factor amino-terminal domain, which is imported into the nucleus and activates the expression of target genes. The UPR has a primary function in adaptive response in order to restore homeostasis and promote cell survival, but depending on the duration and intensity of the stress, a switch can induce cell death to get rid of the damaged cells.
Figure 1. The different UPR effectors and their modes of action. In the basal state, the three UPR effector transmembrane proteins (PERK, ATF6, and IRE-1) are maintained inactive through their interaction with the protein chaperone BiP. The accumulation of misfolded proteins in the ER lumen results in dissociation of BiP and activation of UPR. (1) PERK dimerizes and phosphorylates the eIF2α subunit, leading to a global inhibition of translation initiation. Specific mRNA subsets, containing cis-acting elements in their 5′UTR, such as uORF and IRES, escape translational inhibition triggered by eIF2 phosphorylation. (2) IRE-1 initiates an unconventional splicing of XBP-1 mRNA. IRE1α cleaves Xbp1u mRNA within two stem-loop structures, leading to excision of 26 nucleotides. Subsequent ligation of the Xbp1 mRNA by the tRNA ligase RTCB results in a frame shift and allows the translation of the active transcription factor XBP1s, which is imported into the nucleus and activates the expression of target genes. IRE1α mediates also the degradation of some RNAs (this mechanism has been called RIDD for Regulated Ire1-Dependent Decay). (3) BIP dissociation from ATF6 exposes its Golgi Localization Signal. ATF6 is translocated to the Golgi apparatus where proteolysis releases its transcription factor amino-terminal domain, which is imported into the nucleus and activates the expression of target genes. The UPR has a primary function in adaptive response in order to restore homeostasis and promote cell survival, but depending on the duration and intensity of the stress, a switch can induce cell death to get rid of the damaged cells.
Biomolecules 11 00199 g001
Figure 2. Schematic of the HSC differentiation hierarchy in normal hematopoiesis. HSC, Hematopoietic stem cells; LT-HSCs (Long-Term Hematopoietic Stem Cell) are able to generate new LT-HSC or to differentiate into ST-HSC (Short-Term Hematopoietic Stem Cell) then into MPPs (MultiPotent Progenitors) with reduced self-renewal capacity. Downstream of MPPs, a strict separation takes place between the myeloid (CMP, Common Myeloid Progenitors) and lymphoid (CLP, Common Lymphoid Progenitors) lineages. CMP then produce MEPs (Megakaryocyte–Erythrocyte Progenitors), which differentiate into platelets and erythrocytes, and GMPs (Granulocyte-Macrophage Progenitors) produce granulocytes (neutrophils, eosinophils, and basophils) and macrophages. In the lymphoid lineage, the CLPs then produce T and B lymphocytes and natural killer cells. The whole hematopoietic differentiation process is tightly regulated by a number of intrinsic and extrinsic factors, like cytokines and transcription factors.
Figure 2. Schematic of the HSC differentiation hierarchy in normal hematopoiesis. HSC, Hematopoietic stem cells; LT-HSCs (Long-Term Hematopoietic Stem Cell) are able to generate new LT-HSC or to differentiate into ST-HSC (Short-Term Hematopoietic Stem Cell) then into MPPs (MultiPotent Progenitors) with reduced self-renewal capacity. Downstream of MPPs, a strict separation takes place between the myeloid (CMP, Common Myeloid Progenitors) and lymphoid (CLP, Common Lymphoid Progenitors) lineages. CMP then produce MEPs (Megakaryocyte–Erythrocyte Progenitors), which differentiate into platelets and erythrocytes, and GMPs (Granulocyte-Macrophage Progenitors) produce granulocytes (neutrophils, eosinophils, and basophils) and macrophages. In the lymphoid lineage, the CLPs then produce T and B lymphocytes and natural killer cells. The whole hematopoietic differentiation process is tightly regulated by a number of intrinsic and extrinsic factors, like cytokines and transcription factors.
Biomolecules 11 00199 g002
Figure 3. Activation of UPR signaling in leukemia. Different mechanisms of ER stress activation have been reported in leukemia, which include (epi)genetic modifications and genomic instability (e.g., mutations, translocations, hypomethylation), oncogenic signaling, and metabolism rewiring due to a high proliferation in blasts. Microenvironment is also a well-known source of ER stress (e.g., hypoxia) that contributes to UPR activation.
Figure 3. Activation of UPR signaling in leukemia. Different mechanisms of ER stress activation have been reported in leukemia, which include (epi)genetic modifications and genomic instability (e.g., mutations, translocations, hypomethylation), oncogenic signaling, and metabolism rewiring due to a high proliferation in blasts. Microenvironment is also a well-known source of ER stress (e.g., hypoxia) that contributes to UPR activation.
Biomolecules 11 00199 g003
Table 1. Consequences of UPR induction by drugs in leukemic cells. For each drug, the chemical nature, the pubchem compound ID, and the molecular target are provided when available. The type of leukemia on which the work was carried out was provided as well as the inferred or demonstrated role of UPR activation. The involved effectors are also indicated, when they have been identified (n.d.: not determined).
Table 1. Consequences of UPR induction by drugs in leukemic cells. For each drug, the chemical nature, the pubchem compound ID, and the molecular target are provided when available. The type of leukemia on which the work was carried out was provided as well as the inferred or demonstrated role of UPR activation. The involved effectors are also indicated, when they have been identified (n.d.: not determined).
MoleculeChemical NaturePubchem Compound CIDTargetType of LeukemiaProposed Role of ER Stress/UPR ActivationMainly Implicated Pathways/EffectorsRef.
BIX-01294 25150857Histone methyltransferase G9AAMLAdaptivePERK (prosurvival, via NRF2)[170]
SCH727965 (Dinaciclib) 46926350CDKs 1,2,5 and 9AML, CML, T-ALLAdaptiveXBP1s[171]
Ski, ROMe 16760659spingosine kinases 1 and 2T-ALLAdaptiveunclear[172]
MDA-7/IL-24)cytokine-n.d.AML, APLAdaptiveGRP78/Bip, IRE1α, GADD34[173]
sorafenibmultikinase inhibitor216239MEK/ERK pathwayU937 cell lineAdaptivePERK[174]
Digoxinecardiac glycoside2724385Na+/K+ ATPaseK562 (erythroleukemic) and THP-1 (acute monocytic leukemia) cell linesAdaptivePERK, IRE1α[175]
Shikoninnaphthoquinone479503pyruvate kinase-M2 (PKM2), proteasome inhibition, NFkB, Thioredoxin reductase (TrxR1)HL60AdaptiveERP57 and Calreticulin[176]
3-deazaneplanocin A (DZNeP)cyclopentenyl analog of 3-deazaadenosine73087histone methyltransferaseMV4-11, MOLM-14, Mono-Mac-1, THP-1, HL60 and KG-1 AML cell linesAdaptiveGRP78, GRP94, PERK, PDIA isoform 3, 4, and 5[177]
wolfberry phytochemicalsn.d.-n.d.Jurkat cell lineAdaptiveall UPR pathways[178]
Imatinib 5291tyrosine kinases inhibitorCML/LAMA-84 CML cell line and murine myeloid progenitor primary cellsThe constitutive activation of PERK in CML cells protects from imatinib treatmentPERK[162]
Metformin 4091multiple, see PMID: 28776086T-ALL, B-ALLSwitch form adaptive to terminalIRE1α, CHOP[179]
Nilotinib + MKC8866 644241Tyrosine kinases (Nilotinib); IRE1α (MKC8866)ALL (Ph+)Switch form adaptive to terminalIRE1α (cytoprotective); PERK and ATF6α (cytotoxic)[169]
2-deoxy-D-glucoseglucose analog108223n.d.ALL Cell LinesSwitch form adaptive to terminalGRP78/Bip, CHOP[180,181]
Selenitesodium selenite24934n.d.NB4 cell line (APL)Switch form adaptive to terminalPERK/eIF2α/ATF4[182]
Asperulosideiridoid glycoside84298n.d.cell lines HL60 and U937, primary leukemic cellsTerminal: apoptosis inductionall UPR pathways[183]
JA3 & JA7Aldehyde biphenyl chalcones134820953n.d.(AML); T-ALL; CMLTerminal: immunogenic apoptosis-like cell deathCHOP; PERK[184]
Oprozomibtripeptide analog of carfilzomib25067547immunoproteasome subunit β5i/LMP7 (ubiquitin–proteasome pathway)CMLTerminal: apoptosis inductionPERK, IRE1α (via ASK/JNK/Bim)[185]
VAS3947given in [186] fig 1a7471335NADPH oxidasesAMLTerminal: apoptosis inductionIRE1α, PERK[186]
Arsenic trioxide + Gilteritinib 14888/49803313FLT3 (for Gilteritinib)AML (FLT3-ITD)Terminal: apoptosis inductionIRE1α[187]
GSK-J4 71729975H3K27me3 demethylaseAMLTerminal: apoptosis inductionPKCα; Bcl2 phosphorylation[188]
CXL1464H-chromene derivative- AML (or APL): HL60; CMLTerminal: apoptosis inductionPERK, IRE1α, ATF6α[189]
FF-10501given in paper124343inosine monophosphate deshydrogenaseAMLTerminal: necrotic cell deathCHOP[190]
Retinoic acid+Tunicamycin+ arsenic trioxide 444795/11104835n.d.AMLTerminal: Cytotoxic UPRCHOP, XBP1s[191]
[Retinoic acid or arsenic trioxide] + tunicamycin 444795/11104835n.d.APLTerminal: Cytotoxic UPR [192]
MIM1 and UMI-77 135691163/992586n.d.AML; T-ALLTerminal: Cytotoxic UPRNOXA[193]
PFRpeptide-n.d.AMLTerminal: necroptosis [193]
GenisteinIsoflavone5280961n.d.AML or APL (HL60 cell line)Terminal: Cytotoxic UPR [194]
CamalexinPhytoalexin (structure given in paper)636970n.d.AMLTerminal: apoptosis inductionPERK, CHOP[195]
Ibrutinib (PCI-32765) 24821094Bruton’s tyrosine kinaseB-ALLTerminal: apoptosis inductionATF4; CHOP[196]
OT-55bis-coumarine derivative-n.d.CMLTerminal: Immunogenic cell death induction; apoptosis inductionnot well documented[197]
RS-F3fistularin-3 stereoisomer- AMLTerminal: Cytotoxic UPR (assumed)PERK; XBP1s; CHOP[198]
Bardoxolone methyl (CDDO-Me)triterpenoid400769Nrf2 and NF-κBChronic myeloid leukemia, K562 cell lineTerminal: apoptosis inductionPERK, IRE1α, CHOP[199]
3-O-trans-p-coumaroyl-alphitolic acid (3OTPCA)triterpenoid-n.d.U937, Molt-4 and Jurkat cell lines.Terminal: apoptosis inductionXBP-1 and CHOP[200]
Nelfinavir 64143HIV protease inhibitorsT-ALL, B-ALL, and AML; CLL primary leukemic cellsTerminal: apoptosis induction; In CLL, contributes to the induction of cell death in choroquine treated cellsCHOP[201,202]
CB-50831-[4-(benzylamino)-5H,7H,8H-pyrano[4,3-d]pyrimidin-2-yl]-2-methyl-1H-indole-4-carboxamide439268Valosin-Containing Protein/p97B-ALL Cell Lines (BALL1, REH, NALM6, OP1, ALL-PO, 697, RS4;11, BV173, SEM, and SUPB15)Terminal: apoptosis inductionall UPR pathways[203]
Tunicamycin ± Quizartinib (AC220)AC22011104835 + 24889392FLT3AML (FLT3-ITD)Terminal: apoptosis inductionPERK, CHOP[204]
Cryptotanshinonelipophilic diterpene quinone160254n.d.CCRF-CEM cell line (ALL)Terminal: apoptosis inductionIRE1α-XBP1, PERK-eIF2α-ATF4[205]
Oxalicumone Adihydrothiophene-condensed sulfur chromone90676613n.d.KG-1a, HL60, U937, and K562 cell lines (AML)Terminal: apoptosis inductionIRE1α-XBP1, PERK--CHOP[206]
Pevonedistat (MLN4924)adenosine sulfamate analog16720766NEDD8-activating enzymeT-ALL (CCRF-CEM, Jurkat) and B-ALL (REH, NALM6, SupB15) cell linesTerminal: apoptosis inductionall UPR pathways[166]
Carfilzomib (PR-171)tetrapeptide epoxyketone11556711ubiquitin–proteasome pathwayCLL MEC1 and MEC2 cell lines and primary leukemic cellsTerminal: apoptosis inductionATF4, CHOP[207]
Miltironeabietane-type norditerpenoid quinone160142n.d.Jurkat, U937, AML and ALL primary leukemic cellsTerminal: apoptosis inductionPERK[208]
Arsenic trioxide 14888n.d.NB4 cell line (AML)/CMLTerminal: apoptosis inductionIRE1α-XBP1/GRP78/Bip, CHOP, Xbp1 (unspliced…)[209,210]
JPH203O-[(5-Amino-2-phenyl-7-benzoxazolyl)methyl]-3,5-dichloro-L-tyrosine dihydrochloride24853505LAT1 (L-type amino-acid transporter 1)Ke37, DND41, Sil-ALL, Peer, Molt-16, Jurkat and SupT1 T-ALL cell linesTerminal: apoptosis inductionCHOP[211]
Wogonin5,7-dihydroxy-8-methoxyflavone5281703n.d.HL-60 cell line.Terminal: apoptosis inductionall UPR pathways-CHOP[212]
Farnesolacyclic sesquiterpene alcohol445070n.d.Molt4 T-ALL cell lineTerminal: apoptosis inductionPERK-eIF2α-ATF3/4[213]
APO866(E)-N-[4-(1-benzoylpiperidin-4-yl)butyl]-3-pyridin-3-ylprop-2-enamide6914657nicotinamide phosphoribosyltransferase (NAMPT)OCI/AML2, OCI/AML3, HL-60, HEL, KG1a, SET1, MV4-11, MEC.1, MEC.2, LAMA-84 cell lines and B-CLL and AML primary leukemic cellsTerminal: apoptosis inductionIRE1α-CHOP[214]
Bortezomibboronic acid387447proteasome (26S)NB4 cell line (APL)Terminal: apoptosis inductionNd[215]
CX-49455-(3-chloroanilino)benzo[c][2,6]naphthyridine-8-carboxylic acid24748573casein kinase 2T-ALL cell lines and primary cellsTerminal: apoptosis inductionGRP78/BIP-IRE1α-CHOP[216]
compound 3 (Pyrimidine analogue)1-(5,5,5-trichloro-2-methoxy-4-oxopenten-2-yl)-4-trichloromethyl-pyrimidin-2(1H)-one-n.d.L1210, CEM, JURKAT cell line (ALL)Terminal: apoptosis inductionCHOP and caspase-12[217]
R7, R13Naphtylchalcones n.d.murine lymphoblastic leukemiaTerminal: apoptosis induction (assumed)CHOP[218]
S1 (BH mimetic) APLTerminal: cytotoxic through NOXA inductionPERK; XBP1s, NOXA[219]
Gossypol (BH3 mimetic)polyphenol3503phospholipase A2AML, APLTerminal: cytotoxic through NOXA inductionPERK; NOXA[220]
Cariporide 151172Na + H+ exchanger 1 (NHE1)CML, APL, T-ALLTerminal: sensitizes to extrinsic, TRAIL-induced, apoptosisCHOP[221]
Auranofin 6333901thioredoxin reductaseCLLTerminal: contributes to the induction of cell death in treated cellsPERK; XBP1s, CHOP[222]
[Cu(thp)4][PF6]phosphine copper(I) complex n.d.B-ALLTerminal: apoptosis inductionXbp1s, CHOP[223]
Z-Leu-Leu-Nle-CHOleupeptin analog-γ-SecretaseCLL primary leukemic cellsTerminal: apoptosis inductionIRE1α, CHOP[224]
curcuminDiferuloylmethane969516n.d.WEHI-3 myelomonocytic leukemia cell line/NB4 and UF-1 APL cell lines/HL60 cell lineTerminal: apoptosis inductionIRE1α, ATF6α, CHOP/PERK, CHOP, ASK[225,226,227]
LQB 118pterocarpanquinone46233300n.d.K562 and Jurkat cell linesTerminal: apoptosis induction (assumed)caspase 12[228]
FlavopiridolFlavonoid alkaloid5287969CDKs inhibitorCLL primary leukemic cellsTerminal: contributes to the induction of cell death in choroquine treated cellsIRE1α/XBP1 and CHOP[229]
Safrole 5144 AML (HL-60)Terminal: apoptosis induction (assumed)ATF6α, CHOP[230]
Clofibrate 2796peroxisome proliferator-activated receptor (PPAR) alphaT-ALLTerminal: apoptosis induction (assumed)ASAPK/JNK[231]
Abnobaviscum F ®Mitletoe aqueous extract135343633n.d.CMLTerminal: contribution to the induction of cell death in treated cells (assumed)GRP78/Bip, CHOP[232]
MJ-29Quinazolinone-n.d.murine myelomonocytic leukemiaTerminal: apoptosis inductionGRP78/Bip, CHOP, PERK[233]
Imatinib (STI571) 5291BCR-ABL tyrosine kinase Terminal: apoptosis inductionnot well documented[234]
glycyrrhizic acid 14982n.d.murine myelomonocytic leukemiaTerminal: contributes to the induction of cell death in treated cellsGRP78/Bip, CHOP[235]
Gypenosides -n.dHL-60 AML cell lineTerminal: apoptosis inductionATF6α and ATF4[236]
AICAr (+ methotrexate)5-aminoimidazole-4-carboxamide (AICA) riboside266934n.dNalm6 and CCRF-CEM cell lines (ALL)Terminal: apoptosis inductionCHOP (C/EPB homologous protein)[237]
Emodin6-methyl-1,3,8-trihydroxyanthraquinone3220n.d.WEHI-3 murine myelomonocytic leukemia cell lineTerminal: apoptosis induction (assumed)n.d.[238]
Syrbactinazamacrocyclic product-proteasome (26S)REH ALL cell lineTerminal: apoptosis inductionCHOP (C/EPB homologous protein)[239]
ABT-737 and GX15-070BH3 mimetics11228183/46930997BCL2 family proteinsJurkat, NB4 and K562 cell linesTerminal: Cytotoxic UPRATF4, ATF3, CHOP and NOXA,[240]
NPB001-05n.d.-BCR-ABLK562 cell lineTerminal: apoptosis induction (assumed)not well documented[241]
Ras inhibitor farnesylthiosalicylic acid (FTS, Salirasib)2-[[(2E,6E)-3,7,11-trimethyl-2,6,10-dodecatrien-1-yl]thio]-benzamide5469318RASK562 cell lineTerminal: Cytotoxic UPRnot well documented[242]
PYZD-44093,5-dioxopyrazolidine compound, 1-(3-chloro-4-fluorophenyl)-4-[(5-nitro-2-furyl)methylene]-3,5-pyrazolidinedione60111983ubiquitin-activating enzyme UBA1K562, NB4, THP1, and U937 cell lines and AML primary leukemic cellsTerminal: apoptosis inductionPERK, CHOP, ATF4[243]
Korbazoln.d.-n.d.CLL primary leukemic cellsTerminal: apoptosis induction (assumed)n.d.[244]
Polymethoxyflavone tangeretin (TAN)Flavonoids-n.d.K562 cell lineTerminal: apoptosis inductionIRE1α, PERK, CHOP[245]
Shiga toxine type 1 (Stx1)n.d.-ribosomes (protein synthesis)THP-1 cell lineTerminal: apoptosis inductionCHOP, TNF-related apoptosis-inducing ligand (TRAIL), DR5 and calpain[246]
Eicosapentaenoic acid 446284n.d.HL60 (AML or APL)Terminal: apoptosis induction (assumed)PERK[247]
Xanthohumolprenylated chalcone639665n.d.CLL (patient samples)Terminal: apoptosis inductionPERK, CHOP[248]
Tunicamycin (UPR inducer) 11104835N-acetylglucosamine phophotransferaseAML (U937 and HL60)Terminal: cytotoxic through induction of lysosomal apoptotic pathwayGRP78/Bip, CHOP[249]
arsenic sulfide[As4S4 (AS)]61569n.d.BCR/ABL-positive K562 cell lineTerminal: apoptosis inductionnot well documented[250]
Fenretinidesynthetic retinoid derivative (related to vitamin A)5288209n.d.NB4, U937 and HL60 cell linesTerminal: apoptosis inductionPERK/eIF2α-CHOP (C/EPB homologous protein)[251,252]
PABA/NOO2-[2,4-dinitro-5-(N-methyl-N-4-carboxyphenylamino)phenyl]1-(N,N-dimethylamino)diazen-1-ium-1,2-diolate-PDIHL60 cell lineTerminal: Cytotoxic UPRCHOP (C/EPB homologous protein)[253]
alkyl gallate and gallamide derivatives -n.d.HL60 cell lineTerminal: apoptosis inductionnot well documented[254]
Trichosanthintype I ribosome-inactivating protein596174RibosomesHL60 cell lineTerminal: apoptosis inductionCHOP (C/EPB homologous protein)[255]
auraptenemonoterpene coumarin ether1550607n.d.Jurkat cell lineTerminal: apoptosis inductionCaspase 8[256]
4-hydroxybenzylretinonefenretinide analogue/synthetic retinoid derivative (related to vitamin A)-n.d.HL60 cell lineTerminal: Cytotoxic UPRCHOP (C/EPB homologous protein)[257]
tipifarnib combined with bortezomibquinolone and boronic acid159324/387447Farnesyltransferase Inhibiteur and 26 s proteasome inhibitorKG-1, and U937 cell linesTerminal: Cytotoxic UPRnot well documented[258]
AEBSF4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride186136serine protease inhibitorNB4 cel lineTerminal: Cytotoxic UPRnot well documented[259]
Thapsigargin (UPR inducer)sesquiterpene lactone446378sarco/endoplasmic reticulum Ca++ ATPaseK562 cell lineTerminal: apoptosis inductionnot well documented[260,261]
arsenic trioxide (ATO) + kinase inhibitor imatinib mesylate (STI571) 14888/5291BCR-ABL tyrosine kinaseK562 cell line and CML primary leukemic cellsTerminal: apoptosis inductionnot well documented[262]
Tetrocarcin-A 54681516n.d.CLL/T-ALLTerminal: apoptosis inductionnot well documented[263,264]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Féral, K.; Jaud, M.; Philippe, C.; Di Bella, D.; Pyronnet, S.; Rouault-Pierre, K.; Mazzolini, L.; Touriol, C. ER Stress and Unfolded Protein Response in Leukemia: Friend, Foe, or Both? Biomolecules 2021, 11, 199. https://doi.org/10.3390/biom11020199

AMA Style

Féral K, Jaud M, Philippe C, Di Bella D, Pyronnet S, Rouault-Pierre K, Mazzolini L, Touriol C. ER Stress and Unfolded Protein Response in Leukemia: Friend, Foe, or Both? Biomolecules. 2021; 11(2):199. https://doi.org/10.3390/biom11020199

Chicago/Turabian Style

Féral, Kelly, Manon Jaud, Céline Philippe, Doriana Di Bella, Stéphane Pyronnet, Kevin Rouault-Pierre, Laurent Mazzolini, and Christian Touriol. 2021. "ER Stress and Unfolded Protein Response in Leukemia: Friend, Foe, or Both?" Biomolecules 11, no. 2: 199. https://doi.org/10.3390/biom11020199

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

Article Metrics

Back to TopTop