Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2020 Aug 1;10(8):2226–2241.

Understanding the roles of stress granule during chemotherapy for patients with malignant tumors

Yuting Zhan 1, Haihua Wang 1, Yue Ning 1, Hongmei Zheng 1, Sile Liu 1, Yang Yang 1, Ming Zhou 2, Songqing Fan 1
PMCID: PMC7471355  PMID: 32905441

Abstract

The assembly of stress granules (SGs) is a conserved mechanism to regulate protein synthesis under cell stress, where the translation of global protein is silenced and selective protein synthesis for survival maintains. SG formation confers survival advantages and chemotherapeutic resistance to malignant cells. Targeting SG assembly may represent a potential treatment strategy to overcome the primary and acquired chemotherapeutic resistance and enhance curative effect. We conduct a comprehensive review of the published literatures focusing on the drugs that potentially induce SGs and the related mechanism, retrospect the relationship between SGs and drug resistance related proteins, illuminate the regulated pathways and potential targets for SG assembly, and discuss future directions of overcoming the resistance to chemotherapy.

Keywords: Stress granule, G3BP1, chemotherapy, chemoresistance, mTOR signaling

Introduction

Eukaryotic cells are formed of many compartments or organelles to separate or concentrate biological progress. Taking protein translation as an example, translation initiates in the cytosol for secretory or integral membrane proteins, ribosomes containing mRNAs are recruited to the endoplasmic reticulum (ER) membrane. Once the translation are complete, membrane proteins will be shifted and anchored within the phospholipid bilayer, while secretory proteins may undergo folding and modifications in the ER and be released by the chaperones and packaged by Golgi apparatus for vesicle trafficking [1-3]. ER and Golgi apparatus are membranous organelle and the ribosome is non-membranous. The membranous or non-membranous organelles are playing important roles in protein translation, whereas non-membranous compartments can’t be neglected in the suspended translation process especially for cells exposed to the stress.

Cancer cells are exposed to adverse conditions in the tumor microenvironment such as nutrient deprivation, hypoxia, DNA damage, oxidative stress, inflammation, reduced PH, immune attack and radical or chemical treatment, which compels malignant cells to make adaptive changes to ensure survival [4,5]. When confronted with stress, one highly conserved mechanism is reducing global protein synthesis and maintaining selective protein synthesis that of the essence for cell survival [6-8]. Stress granules (SGs), one kind of non-membranous compartment in the cytoplasm, are assemblies of untranslating messenger ribonucleoproteins (mRNPs) that form from mRNAs stalled in translation initiation [9]. Since no membrane-like structure, interactions between protein-and-protein as well as protein-and-RNA are significant. Besides mRNPs, many other components are also involved in the assembly of SGs including RNA-binding proteins (such as G3BP1 [10-12], IMP1 [10], TIA1 [13], et al.), translation initiation factors (such as eIF3 [14], eIF4A/B [14,15], eIF4E [16], eIF4G [14] , et al.), poly-A-binding protein (such as Pab1 in saccharomyces [17], PABP1 [18], et al.) and ribosomal subunits (such as 40S subunits [11,12]). In most cases, the formation of SGs is requiring the phosphorylation of the translation initiation factor eIF2α [19]. Under steady-state conditions, eIF2/tRNAi Met/GTP ternary complex can bind initiator tRNAi Met to the 40S ribosomal subunit in a GTP-dependent manner. Adverse conditions activate the eIF2α kinases and lead to phosphorylation of eIF2α, which damage the ternary complex and impair translational initiation, following the formation of SGs [5]. Phosphorylation of eIF2α on serine 51 can be activated by a family of four kinases, heme-regulated inhibitor (HRI), protein kinase R (PKR), PKR-like endoplasmic reticulum kinase, (PERK) and general control non-depressible 2 (GCN2) [20-22]. All four kinases have catalytic domain, and are supposed to be activated by homodimerization and autophosphorylation [23]. Each kinase can be activated by a specific stress. HRI is activated during heme deficiency [24]; PKR is activated by viral infection [25,26]; PERK is activated during ER stress [27]; GCN2 is activated under amino acid deprivation [28]. Different stimulus can cause different intensity of eIF2α phosphorylation. NaCl, ultraviolet light and thapsigargin cause strong eIF2α phosphorylation; cold shock, H2O2, heat shock, low glucose, arsenate, and histidinol cause moderate phosphorylation; polyinosinic polycytidylic acid, anoxia, and serum starvation cause mild phosphorylation [21] (Figure 1). In some cases such as response to mammalian orthoreovirus, the formation of SGs is eIF2α independent [29]. Besides, in the brain ischemia-reperfusion process, SG formation is correlated with the decreased expression of the cap-binding protein eIF4E and the eIF4B [30]. Selenite can induce SGs formation via eIF4E-binding protein 1 (4EBP1)-mediated inhibition of translation initiation [31].

Figure 1.

Figure 1

The assembly of stress granules in the phosphorylation of eIF2α dependent manner. Under steady-state conditions, eIF2/tRNAi Met/GTP ternary complex can bind initiator tRNAi Met to the 40S ribosomal subunit in a GTP-dependent manner. Adverse conditions activate the eIF2α kinases (HRI, PKR, PERK and GCN2) and lead to phosphorylation of eIF2α, which damage the ternary complex and impair translational initiation, following the formation of SGs. SGs are assemblies of untranslating messenger ribonucleoproteins that form from mRNAs stalled in translation initiation, also containing RNA-binding proteins, translation initiation factors, poly-A-binding protein and 40S ribosomal subunits.

SGs are non-membrane bound cytoplasmic entities, and “Core first” model and “liquid-liquid phase separations (LLPS) first” model have been established to explain the assembly of SGs. “Core first” model, the traditional concept, is a process starting to untranslating mRNAs with bound SG-nucleating proteins (such as TIA1, G3BP1, TTP, FMRP, CAPRIN1 et al.) oligomerize into stable cores, and the outer shell forms later [32,33]. The other concept of SGs formation is called “LLPS first” model, which thinks SG formation before the core concentrate. In this view, the increasing pool of untranslated mRNAs bound by proteins containing intrinsically disordered protein regions (IDRs), firstly lead to the formation of a LLPS based on IDR-IDR interactions. And the cores assemble following with the increased local concentration of its components [34].

SGs assembly is a conserved cellular response to minimize stress-related damage and promote cell survival. The aberrant assembly or disassembly of SGs is believed to participate in neurodegenerative disorders, ischemia-reperfusion process, virus infections and cancer initiation or development. Various chemotherapy drugs can modulate SG formation and dynamics, in the meantime, SGs, as the signaling center, are promising targets for cancer treatment. In this review, we summarize the clinical drugs for inducing SGs assembly and their related mechanism, as well as the potential roles of SGs in cancer treatment, in the interest of providing new perspectives for overcoming chemotherapy resistance.

Stress granules can be induced by chemotherapy drugs

Cancer treatment is now multidisciplinary. In theory, it is potential to cause SGs assembly by any chemotherapeutic drug that influence the translation process or targeting translation element in cells (Figure 2).

Figure 2.

Figure 2

Stress granules can be induced by chemotherapy drugs. Platinum induce reactive oxygen species and lead to SGs assembly. PKR protein can be activated by 5-Fluorouracil, inducing phosphorylation of eIF2α and cell death by apoptosis. Arsenic trioxide, vinorelbine and bortezomib can cause phosphorylation of eIF2α by activating PKR, PERK and HRI respectively.

Platinum and platinum-containing drugs

Platinum, the most effective agent for nearly all types of malignant tumor, has been linked its effect to the ability of influencing the purine bases on the DNA, interfering with DNA repair mechanisms, causing DNA damage, blocking cell division and inducing apoptosis in cancer cells [35]. Not only damaging DNA directly, platinum also induce reactive oxygen species (ROS) in cells and cause oxidative stress [36,37]. Oxidative stress is one of the conditions that induce SG assembly. ROS, such as H2O2, is routinely used as inducer for oxidative stress or SGs [33,38]. Ataxin-2-like (ATXN2L) is a regulator of SGs and processing bodies. ATXN2L overexpression induces the formation of SGs, while the reduced ATXN2L affects the size and number of SGs [39]. ATXN2L is found upregulated in gastric cancer tissue and indicated adverse prognosis for overall survival and recurrence. Oxaliplatin is proved to promote ATXN2L expression and SG assembly. The oxaliplatin-resistant cell lines present with elevated ATXN2L levels, while silencing ATXN2L can reverse the oxaliplatin resistance by increasing ROS production and apoptosis [40]. In contrast to oxaliplatin, cisplatin fails to induce immunogenic tumor cell death, which may attribute to its incapacity to translocate calreticulin from the lumen of the ER to the cell surface [41]. It is indicated that cisplatin is unable to activate the PERK-dependent phosphorylation of eIF2α in U2OS cells (osteosarcoma cell), and fails to stimulate the formation of SGs. When combined cisplatin and thapsigargin (an inhibitor of the sarco/ER Ca2+-ATPase, does not stimulate calreticulin exposure), phosphorylation of eIF2α and SGs can be detected [42]. Certainly, the ability of inducing SGs depends on cell types, cisplatin may lead to SGs assembly in malignant glioma cells, and impairment of SG assembly may sensitize cells to cisplatin [43].

In fact, it is difficult to delimitate the advantageous or disadvantageous roles for SGs in cancer treatment. After cisplatin treatment, more dead cells were found in G3bp1-knockdown cells (less SGs formation) compared with controls, accompanied by increases in cleaved/active caspase-3. SGs are formed to protect proximal tubular cells under adverse condition [44]. The combination of cisplatin and thapsigargin may promote phosphorylation of eIF2α and SGs formation, enhance immunogenic cell death [42], which drives efficient antitumor effects [45]. The above studies show that the formation of SGs may play positive roles in cancer treatment, for protecting proximal tubular cells or increasing the antitumor effects. However, some studies indicate that oxidative stress and SGs formation facilitate cancer cells to acquire chemoresistance, which is negative for cancer treatment [40,43].

5-Fluorouracil (5-FU)

The mechanism of cytotoxicity of 5-FU has been attributed to the inhibition of the nucleotide synthetic enzyme thymidylate synthase (TS), and to the misincorporation of 5-FU metabolites into DNA and RNA. When 5-FU metabolites incorporate into RNA, the processing and maturation of rRNA, tRNA and snRNA are all influenced [46,47]. Phosphorylation of eIF2α may damage eIF2/tRNAi Met/GTP ternary complex and impair translational initiation, leading to the formation of SGs [5]. It is reported that PKR protein can be activated by 5-FU, inducing phosphorylation of eIF2α and cell death by apoptosis [48]. Clinically, PKR and its regulator, the non-coding RNA pre-miR-886 (nc886), are established to evaluate the patients’ prognosis and response to 5-FU-based chemotherapy. Higher levels of nc886 predicts better response to treatment, and the cases lacking PKR location in the nucleolus show a positive relationship with 5-FU-based chemotherapy [49]. Interesting, PKR has been identified as the key target for 5-FU promoting apoptosis; however, the active PKR also leads to phosphorylation of eIF2α and induce SGs assembly, thereby assisting tumor cells overcome 5-FU-induced cytotoxicity and leading to chemoresistance [50]. 5-FU-induced SGs contain RACK1, a promoter for cell apoptosis, and the sequestration of RACK1 to SGs may suppress the stress-responsive MAPK pathways therefore inhibiting apoptotic cascades and inducing resistance to chemotherapy [50-52]. It seems to be contradictory that increased expression of PKR is associated with better clinical outcome for lymph node negative rectal cancer patients who have received post-operative chemoradiation based on 5-FU [53]. We recommend readers to keep a watchful eye on tumor load, the study of Ortega-García MB et al. intakes colon metastatic cancer patients with unresectable lung or liver metastases [49], while Kwon HC and his colleagues observe lymph node negative rectal cancer patients [53]. Despite multiple studies of PKR, the exact role in cancer biology and integrated stress response (ISR) remains controversial. On the one hand, PKR can induce apoptosis via caspase-8 and caspase-9 pathway [54]; on the other hand, PKR may lead to phosphorylation of eIF2α and promote SGs assembly, leading to chemoresistance.

Arsenic trioxide (ATO) and sodium arsenite (SA)

Low-dose ATO, an agent that induces oxidative stress and interferes with protein translation, is always combined with all-trans-retinoic acid (ATRA) to treat acute promyelocytic leukemia [55], while large-dose ATO or SA is the most common reagent for inducing SGs [21]. In vitro, the ATO concentration for inducing cell differentiation is always less than 1 uM [56,57], for studying cell apoptosis is between 1 and 5 uM [57-59], for initiating SGs formation is from 100 to 500 uM (about 1 hour) [11,16,60]. The specific concentration and time differs from each experiment, however, the incremental concentration seems to enlighten us that SGs formation is a kind of rapid-response strategy to cope with the lethal attack. When treated with ATO, the SGs formation is PKR and phosphorylation of eIF2α dependent [61]. One of antitumor methods for ATO is the induction of apoptosis, and pretreatment of ER stress inducer may enhance ATO efficiency; activation of p53 can be observed in this process [59,62]. ER stress can be induced by ATO, a mitochondrial toxin, and triggers tumor cells apoptosis involving interplay of ER and mitochondria [63]. However, the lethal stress also urges cells form SGs and strives for ability to survive, leading to resistance to chemotherapy. The overexpression of Musashi-1 (MSI1) can be detected in tumor tissues compared with adjacent normal tissues, and correlated with poor overall survival [64,65]. MSI1 activates PKR/p-eIF2α/SGs axis in response to cytotoxic stress from ATO treatment, and reduces ATO-induced apoptosis in glioblastoma multiforme cells [66]. ATO functions as effective anti-tumor drug for triggering cell apoptosis; however, there are various mechanisms for malignant cells to response to ATO and evade apoptosis, and inducing SGs formation may be a potential synergia for overcome ATO resistance. For example, cells become resistant to SA after repeated SA treatment, which changes in SG biology and the pool of secreted factor, increasing survival response and resulting in chemo-resistance [67].

Phytogenic anticarcinogen: vinca alkaloids and paclitaxel

Microtubule-targeting agents (MTAs) such as paclitaxel and vinca alkaloids are one of the most important chemotherapy drugs available to combat cancer. MTAs influence mitotic spindle formation by interfering microtubule dynamics during mitosis, leading to cell cycle arrest, apoptosis, vascular disrupting to combat cancer [68,69]. It is reported that microtubule dynamic instability favors the assembly of SGs [70]. SGs are induced by SA, vinca alkaloids, the microtubule-depolymerizing drug, may abolish arsenate-induced formation of SGs, while the microtubule-stabilizing drug paclitaxel has the opposite effects [71]. Interestingly, vinca alkaloids (vinorelbine) are the potent inducers of SGs, which is dose- and time-dependent. Vinorelbine promote SG formation in a phospho-eIF2α dependent manner via activation of PERK kinase; and it also promotes dephosphorylation of 4E-BP1 and disrupts eIF4F complex formation. Interestingly, depletion of PERK and/or 4E-BP1 can sensitize cell for vinorelbine and increase cell apoptosis [72].

Treated cells with paclitaxel can induce SGs formation, tubulin is not found in SGs [72]. The specific mechanism for paclitaxel inducing SGs is not known. But to be sure, translation is significantly inhibited during paclitaxel-induced apoptosis in cancer cells, which is involved in elongation factor eEF2, rather than phosphorylation of eIF2α, eIF4G, eIF4E and 4E-BP1, although the decrease of eIF4G, eIF4E and 4E-BP1 expression levels can be detected [73].

Bortezomib

Bortezomib, a peptide boronate inhibitor of the 26S proteasome, is applied in the clinical treatment and observed to improve the prognosis of multiple myeloma and mantle cell lymphoma [74,75]. Bortezomib is proved to result in cell apoptosis, and solid tumor cells are largely refractory to bortezomib [76]. There is no certain mechanism why solid tumor is not sensitive to bortezomib, and SGs formation probably one of the potential explanations. Bortezomib induces the assembly of SGs in cancer cells involving the phosphorylation of eIF2α via HRI activation, causing a reduction of global translation; the disassembly of SGs and the associated translation recovery doesn’t need dephosphorylation of eIF2α [77]. It is reported that inhibition of eIF2α and impairment of SG assembly by knocking down G3BP1 can sensitize gliomas cells to bortezomib [43,78]. Notably, knocking down G3BP1 may significant increase in the apoptotic response to bortezomib with increased expression of caspase-3 [78]. Moreover, Bortezomib induces the localization of CUGBP1-p21mRNA in SGs, while p21 upregulation can promote cancer cell resistance to bortezomib [79]. Given that targeting SGs may enhance the sensitivity to bortezomib, the combination of bortezomib and small molecules seems to be promising method to overcome resistance of bortezomib in solid tumor treatment. It is found psammaplysin F, a kind of marine sponge-derived metabolite, can decrease the number of SGs with the induction of ATO, and enhance the efficacy of bortezomib in HeLa and MCF7MDR cells, which represent a useful strategy to improving drug efficacy [80].

Stress granules contribute to chemotherapy resistance

Stress granules and ABC transporters

The expression of drug resistance-related proteins limits the efficacy of current chemotherapeutic agents. When exposed to stress, eukaryotic cells may selectively shut down the translation of constitutively expressed genes, and simultaneously maintain or enhance the translation of specific stress-induced transcripts [4]. Overexpression of P-glycoprotein, encoded by multidrug resistance 1 (MDR1), is often responsible for multidrug resistance and chemotherapy failure in cancer treatment [81]. It is observed that MDR1 mRNAs do not colocalize with TIA-1 (SG-nucleating protein) and are not sequestered into SGs on arsenite treatment. Interesting, MDR1 and other ER-associated transcripts retain their polyribosomes, tether the transcripts to the ER. On withdrawal of stress, the translational status of MDR1 mRNA recovers more quickly than that of other mRNAs such as ACTB and ATF4 (normally sequestered in SGs [82]), and endows survival priority to tumor cells and induce resistance to chemotheraputic agents [83,84].

Protein kinase C (PKC), a family of serine/threonine kinases, mediates the induction of P-glycoprotein to increase drug resistance in multiple cancers [85]. It is reported that PKCα interacts with G3BP2 and localizes to SGs; downregulation of PKCα delays induction of eIF2α phosphorylation and SG formation [86]. More and deeper studies need to be done to detect the relationship and regulation mechanism of other drug resistance related proteins (such as MRP, BCRP and LRP) and SGs assembly and disassembly in the future.

Stress granules and cell stemness

Cancer stem cells (CSCs), a small amount of cells conferring to the capacity to self-renew, are supposed to partially responsible for chemotherapy resistance, and cancer cells that undergo epithelial-to-mesenchymal transition (EMT) have been shown to acquire stemness and undergo metabolic changes [87,88]. It is reported that knockdown of G3BP1 inhibit the EMT process with the alteration of Cadherins, Vimentin, Snail, Slug, c-Myc, and cyclin D1 [89]. G3BP2 also regulates expression of Oct-4, Nanog and SART3 [90], which are the potential molecular markers for further characterization of CSCs [87]. Meanwhile, Musashi-1, a stemness gene as well as colon and neuronal stem cell marker, triggers the formation of anti-apoptotic SGs with 5-FU and Musashi-1 SGs enhance the chemoresistance of colorectal cancer in turn [91]. In fact, Musashi-1 is proved to be associated with progression and poor prognosis of many kinds of cancers [92,93], and is thought as a novel target in cancer treatment due to its ability to maintain stemness and promote SGs formation [66,94,95].

Stress granules and apoptosis

The therapeutic effects of chemotherapy drugs largely rely on the trigger of a cascade of apoptosis process, and therefore, the assembly of SGs can partially offset the apoptotic functions of chemotherapy drugs and result in chemotherapy resistance. For example, the SG assembly is reduced by bortezomib after knockdown of G3BP1 with increased Caspase-3 activation, enhancing the effects of bortezomib [78]. Inhibiting phosphorylation of eIF2α also promotes apoptosis by reducing SGs assembly [96]. In fact, SGs are the crossroads of apoptosis process and stress response, and regulate type I and type II stress by sequestering RACK1, relevant to hypoxia-induced chemoresistance [52].

Stress granules and autophagy

Autophagy is a double sword in cancer treatment, no matter protective autophagy inhibition or autophagy overactivation may introduce cell death pathway in addition to apoptosis and seem to be effective in combating chemoresistance and radioresistance in malignant tumors [97]. It is validated autophagosomes are associated with clearance of SGs and help cells to survive the stress stimulus. Treated with SA or MG132, Syk is recruited in SGs by Grb7, and the recruitment requires the phosphorylation of Syk on tyrosine. Notably, Syk promotes the clearance of SGs through autophagy, enhancing the ability of cells to survive the stress stimulus [98]. Besides, knockdown of Survivin, an anti-apoptotic molecule, is found to activate autophagy signal, along with the increased number of SGs [99]. It needs further study to reveal whether the activated autophagy signal associated the clearance of SGs and promote malignant cells survive.

Stress granules are the intersections of cell signaling pathways: mTOR signaling pathway as a paradigm

It’s no exaggeration to say that SGs are the intersections of multiple molecules, while regulated signaling pathways are also potent to influence tumor initiation and progression, as well as chemotherapy drugs efficacy (Figure 3).

Figure 3.

Figure 3

The different roles of mTOR signaling in stress granules assembly. A. The activation of mTOR-S6 kinase pathway facilitates malignant progression of malignant tumor, and also promotes SGs assembly. The mTOR-S6 kinase pathway can promote SG assembly in response to mild oxidative stress by eIF2α phosphorylation, and mTOR/4EBP1/eIF4E axis enhances the ability of SGs assembly. B. mTORC1 is a main activator of translation, and thus translation arrest through mTORC1 inhibition may has potential to induce SGs assembly.

The activation of mTOR-S6 kinase pathway facilitates malignant progression of malignant tumor [100], and also promotes SGs assembly. S6K1 influence SGs number and size after mild arsenite treatment, while S6K2 may play a major role in the persistence of SGs, which is mTOR dependent and independent. In mechanism, the mTOR-S6 kinase pathway can promote SG assembly in response to mild oxidative stress by eIF2α phosphorylation [101]. Moreover, mTOR/4EBP1/eIF4E axis enhances the ability of SGs assembly [16]. Interestingly, lack of ataxin-2 (a component of SGs) increases phosphorylation of RPS6 and 4E-BP1 through the PI3K/mTOR pathway [102]. Given that malignant cancer cells require mTOR complex 1 (mTORC1) activity, hyperactivation of mTORC1 may lead to cells apoptosis, mTORC1 activity needs to be balanced in cancer cells [103]. Upon stress, the mTORC1 component raptor can be recruited to SGs, thereby preventing mTORC1-hyperactivation-induced apoptosis [104]. The findings hint that SGs are the intersections of mTOR signaling pathway, which can regulate SGs assembly; its component can be sequestered in SGs; in turn, other SGs component can influence the activation of mTOR signaling pathway. In fact, overexpression of the SG-neucleating protein (G3BP1), SG-formation-regulation protein (YB1) and mTOR signaling member can together predict poor clinical prognosis of non-small cell lung cancer patients [105].

Reversely, another viewpoint suggest that mTORC1 is a main activator of translation, and thus translation arrest through mTORC1 inhibition via nutrient deprivation or small molecule compounds may has potential to induce SGs assembly [22,106]. Controversially, in the model of chronic nutrient starvation, SGs assembly is not directly dependent on decreased mTORC1 activity, and is likely eIF2α phosphorylation dependent [107]. Remarkably, ethanol, inhibition of mTORC1 activity and complex formation, induces the formation of SGs; while INK128, complete inhibition of mTORC1 and mTORC2 activity, suppress the SGs formation in large B-cell lymphoma [108]. Although there is no consistent conclusion for the relationship between mTOR and SGs assembly, the initiation of malignant tumor apparently favor the first perspective. Given that the activation of oncogenic signaling to mTORC1 may facilitate cells grow, meanwhile, mTOR signaling pathway promotes SG assembly to resist adverse conditions and survive from chemotherapy.

In the process of SGs assembly, mTOR is regulated by traditional PI3K/AKT, but also by MAPK signaling pathway. It is reported that PI3K is the main driver of SGs when highly active, and the impact of MAPK/p38 becomes more apparent following PI3K activity declines [109]. Interesting, when SGs assemble, it negatively regulates apoptotic response by segregating RACK1, a scaffold for JNK/MAPK signaling, and thus suppress activation of the MTK1 (a MAPKKK)/JNK/MAPK pathway, which is a potential mechanism to cause chemotherapy [52].

Targeting stress granules assembly may influence the efficacy of chemotherapy drugs

Multiple chemotherapy drugs are potential to cause SGs assembly, and in turn, the formation of SGs may contribute to chemotherapy resistance by a series mechanism. It seems to be a promising target to reverse or avoid resistance of chemotherapy by inhibiting SGs assembly (Figure 4).

Figure 4.

Figure 4

Targeting stress granules assembly may influence the efficacy of chemotherapy drugs. Targeting SG-nucleating proteins, eIF2α and its kinases, other eukaryotic translation initiation factors, and mTOR signaling may influence the SGs formation and the efficacy of chemotherapy drugs.

Targeting SG-nucleating proteins

SG-nucleating proteins, such as G3BP1 and CAPRIN1, are significant for the assembly of SGs and targeting their expression may be a potential strategy to partially reduce chemotherapy resistance caused by SG formation. When silencing G3BP1 mRNA and protein expression in U87 cells, the SG assembly is reduced and the bortezomib-treated cells have a significant increase in the apoptotic response with increased Caspase-3. Moreover, the conditioned culture medium of G3BP1-knocked-down bortezomib-treated cells inhibited angiogenesis compared to control group [78]. It is also reported that CAPRIN1 overexpression protects cancer cells from AS, or docetaxel-induced cell death, while CAPRIN1 knockout sensitizes malignant cells to stress-induced cell death. In the animal model, knockout of CAPRIN1 significantly reduced the growth of tumor xenografts [33].

Targeting eukaryotic translation initiation factors

In most cases, phosphorylation of eIF2α is the initial point of SGs assembly, and thus targeting its phosphorylation process seems to be an effective method to avoid SG-induced survival of malignant cells, enhancing the efficacy of chemotherapy drugs. Cancer cells are exposed to hypoxia in rapidly growing tumors, which increases production of ROS and induces SG formation. Hypoxia induces eIF2α phosphorylation and SG formation, resulting HeLa cells not sensible to cisplatin and paclitaxel. When partially reversing SG assembly by stanolone, HeLa cells are observed more sensible to cisplatin and paclitaxel under hypoxia [96]. Similar enhanced chemotherapeutically-induced cell death can be observed in glioma cells response to bortezomib, cisplatin, and etoposide [43]; MCF7 and HeLa cells to bortezomib [80]; U87 cells to ATO [66]. Silencing PERK, eIF2α kinase, also reverses resistance to ER stress and chemotherapy [110]. These results strongly indicate that Interfering eIF2α and its kinases may be a potential strategy for new co-adjuvant therapies to treat malignant tumors.

Apart canonical eIF2α phosphorylation, other eukaryotic translation initiation factors also facilitate SGs formation such as eIF4A in mutant KRAS cells following exposure to stress-inducing stimuli. It is observed that cell non-autonomous upregulation of SGs by mutant KRAS confers cells the resistance to oxaliplatin [111]. In fact, eIF4A is upregulated in colorectal cancer and predicts poor survival of patients, and knocking-down EIF4A2 sensitizes tumor cells to oxaliplatin treatment [112]. Besides, eIF4E is essential in the selenite-induced SGs assembly [31], and it may enhance efficacy or overcome drug resistance in combination with 5-FU, cisplatin and ATO [113-115]. These findings suggest targeting eIFs as a promising way for cancer treatment.

Targeting mTOR signaling

Although a small amount studies indicate that translation arrest through mTORC1 inhibition may has potential to induce SGs assembly (discussed before) [22,106], more studies verify that mTOR signaling facilitates malignant progression of malignant tumor and also promotes SGs assembly. The later construct the basement of malignant phenotype and chemotherapy resistance of malignant tumors, and provide a feasible scheme for targeting mTOR signaling to enhance chemotherapeutic drug efficacy. It is proved to be influenced on gene translation by ethanol (inhibition of mTORC1 and induction of SGs) or INK128 (complete inhibition of mTORC1 and mTORC2 activity and suppression of SGs) in protein synthesis, cell cycle, proliferation and apoptosis [108]. It is well known that enhanced efficacy can be achieved by inhibition of mTOR, a primary resistance factor [116,117], and whether the assembly or disassembly of SGs participate in this process needs to be further explored. Besides mTOR-S6 kinase pathway, mTOR/4EBP1/eIF4E also participate the assembly of SGs. It is reported that mTORC1-induced eIF4E-eIF4GI interactions facilitate SGs formation, while 4EBP1 inhibits mTORC1-dependent SGs formation by disrupting eIF4E-4GI association. Suppression of SG though depletion of eIF4E and eIF4GI sensitizes cancer cells to bortezomib-mediated apoptosis, involving p21 downregulation [16]. Therefore, there are still lots of work to be done to verify how mTOR signaling influences the chemotherapy resistance in the future.

Conclusions and future perspectives

The assembly of SGs is a conservative strategy for cells to conserve energy and cope with adverse conditions. SGs formation may decrease global proteins synthesis, permitting specific pro-survival mRNA to translate to proteins, and thus ensure cells to survive in numerous stimuli. In fact, SGs widely participate in the physiological and pathological process of cells, playing oncogenic roles via influencing protein translation, proliferation, cell cycle, apoptosis, et al. In response to radiation [118] or chemotherapy drugs, SGs assembly may protect malignant cells to avoid lethal attacks and generate resistance to the treatment. SGs contribute to chemotherapy resistance by various mechanisms, such as facilitating ABC family expression, enhancing stemness of malignant cells and regulating cell apoptosis and autophage. In fact, YBX1 is regarded as the crossroad of P granules, SGs and exosomes, and seems to be a bridge of these non-membrane constructions [119]. Particularly, SGs form spontaneously in the cells with KRAS mutation, which may partially explain the primary drug resistance for some cancer patients. SGs can be induced by various kinds of chemotherapy drugs, in turn, the formation of SGs can decrease therapeutic effects and cause acquired chemoresistance.

The process of protein translation and SGs formation is complicated, and regulated by intrinsic and environmental factors. The activation of eIF2α kinases and phosphorylation of eIF2α is regarded as beginning points of SGs formation, also a few are eIF2α-independent, and other eukaryotic translation initiation factors or mTOR signaling involve. “Core first” model and “LLPS” model are constructed to explain why and how SGs assembly, and either one highlights the irreplaceability of nucleating proteins such as TIA-1 and G3BP1, which highly express in the malignant tissues than the adjacent and indicate the poor prognosis.

In the future, further researches have to be based on the omics analysis, studying the proteomics and metabonomics within and out of SGs. Moreover, SGs are a pool of mRNP, RNA binding proteins, eukaryotic translation initiation factors, poly-A-binding protein and ribosomal subunits, and are the intersections of cell signaling pathways. Therefore, the roles and functions of SGs are complicated and dynamic. More studies about SGs and chemotherapy resistance can be concentrated on the following aspects: (1) clearing the triggering mechanism of SGs coping with chemotherapy drugs, and developing the corresponding targets to avoid SG formation; (2) exploring the possibility of chemotherapy drugs and confirming the dose and time; (3) seeking the mutation of forming SGs spontaneously, and providing the evidence of primary resistance to chemotherapy; (4) studying how the formation of SGs influence the protein expression of proliferation, cell cycle and apoptosis, and partially explaining how acquired resistance to chemotherapy happens; (5) finding the relationship between SGs formation and drug resistance related proteins, such as P-glycoprotein, MRP, BCRP and LRP; (6) facilitating SGs formation in normal tissues to decrease the side effect of chemotherapy; (7) revealing the crosstalk between SGs and other membrane or non-membrane constructions.

Acknowledgements

This work was supported by the National Natural Science Foundations of China (No: 81972838, 81773218, 81472773, 81802791 and 81703009) and The Natural Sciences Foundations of Hunan Province (No: 2018JJ3858 and 2017JJ3457).

Disclosure of conflict of interest

None.

Abbreviations

SG

stress granule

ER

endoplasmic reticulum

mRNP

messenger ribonucleoprotein

HRI

heme-regulated inhibitor

PKR

protein kinase R

PERK

PKR-like endoplasmic reticulum kinase

GCN2

general control non-depressible 2

LLPS

liquid-liquid phase separation

IDR

intrinsically disordered protein region

ROS

reactive oxygen species

ATXN2L

Ataxin-2-like

5-FU

5-Fluorouracil

ISR

integrated stress response

ATO

arsenic trioxide

SA

sodium arsenite

ATRA

all-trans-retinoic acid

MTA

microtubule-targeting agent

MDR1

multidrug resistance 1

PKC

protein kinase C

CSC

Cancer stem cell

EMT

epithelial-to-mesenchymal transition

References

  • 1.Noller HF, Lancaster L, Mohan S, Zhou J. Ribosome structural dynamics in translocation: yet another functional role for ribosomal RNA. Q Rev Biophys. 2017;50:e12. doi: 10.1017/S0033583517000117. [DOI] [PubMed] [Google Scholar]
  • 2.Schwarz DS, Blower MD. The endoplasmic reticulum: structure, function and response to cellular signaling. Cell Mol Life Sci. 2016;73:79–94. doi: 10.1007/s00018-015-2052-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Witkos TM, Lowe M. Recognition and tethering of transport vesicles at the Golgi apparatus. Curr Opin Cell Biol. 2017;47:16–23. doi: 10.1016/j.ceb.2017.02.003. [DOI] [PubMed] [Google Scholar]
  • 4.El-Naggar AM, Sorensen PH. Translational control of aberrant stress responses as a hallmark of cancer. J Pathol. 2018;244:650–666. doi: 10.1002/path.5030. [DOI] [PubMed] [Google Scholar]
  • 5.Anderson P, Kedersha N, Ivanov P. Stress granules, P-bodies and cancer. Biochim Biophys Acta. 2015;1849:861–70. doi: 10.1016/j.bbagrm.2014.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Garre E, Pelechano V, Sánchez Del Pino M, Alepuz P, Sunnerhagen P. The Lsm1-7/Pat1 complex binds to stress-activated mRNAs and modulates the response to hyperosmotic shock. PLoS Genet. 2018;14:e1007563. doi: 10.1371/journal.pgen.1007563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sicari D, Fantuz M, Bellazzo A, Valentino E, Apollonio M, Pontisso I, Di Cristino F, Dal Ferro M, Bicciato S, Del Sal G, Collavin L. Mutant p53 improves cancer cells’ resistance to endoplasmic reticulum stress by sustaining activation of the UPR regulator ATF6. Oncogene. 2019;38:6184–6195. doi: 10.1038/s41388-019-0878-3. [DOI] [PubMed] [Google Scholar]
  • 8.Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A, Gorman AM. The integrated stress response. EMBO Rep. 2016;17:1374–1395. doi: 10.15252/embr.201642195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Protter D, Parker R. Principles and properties of stress granules. Trends Cell Biol. 2016;26:668–679. doi: 10.1016/j.tcb.2016.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Niewidok B, Igaev M, Pereira da Graca A, Strassner A, Lenzen C, Richter CP, Piehler J, Kurre R, Brandt R. Single-molecule imaging reveals dynamic biphasic partition of RNA-binding proteins in stress granules. J Cell Biol. 2018;217:1303–1318. doi: 10.1083/jcb.201709007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kedersha N, Panas MD, Achorn CA, Lyons S, Tisdale S, Hickman T, Thomas M, Lieberman J, McInerney GM, Ivanov P, Anderson P. G3BP-Caprin1-USP10 complexes mediate stress granule condensation and associate with 40S subunits. J Cell Biol. 2016;212:845–60. doi: 10.1083/jcb.201508028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Meyer C, Garzia A, Morozov P, Molina H, Tuschl T. The G3BP1-family-USP10 deubiquitinase complex rescues ubiquitinated 40S subunits of ribosomes stalled in translation from lysosomal degradation. Mol Cell. 2020;77:1193–1205. e5. doi: 10.1016/j.molcel.2019.12.024. [DOI] [PubMed] [Google Scholar]
  • 13.Meyer C, Garzia A, Mazzola M, Gerstberger S, Molina H, Tuschl T. The TIA1 RNA-binding protein family regulates EIF2AK2-mediated stress response and cell cycle progression. Mol Cell. 2018;69:622–635. e6. doi: 10.1016/j.molcel.2018.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ivanov P, Kedersha N, Anderson P. Stress granules and processing bodies in translational control. Cold Spring Harb Perspect Biol. 2019;11:a032813. doi: 10.1101/cshperspect.a032813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jongjitwimol J, Baldock RA, Morley SJ, Watts FZ. Sumoylation of eIF4A2 affects stress granule formation. J Cell Sci. 2016;129:2407–15. doi: 10.1242/jcs.184614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fournier MJ, Coudert L, Mellaoui S, Adjibade P, Gareau C, Côté MF, Sonenberg N, Gaudreault RC, Mazroui R. Inactivation of the mTORC1-eukaryotic translation initiation factor 4E pathway alters stress granule formation. Mol Cell Biol. 2013;33:2285–301. doi: 10.1128/MCB.01517-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Brambilla M, Martani F, Branduardi P. The recruitment of the Saccharomyces cerevisiae poly(A)-binding protein into stress granules: new insights into the contribution of the different protein domains. FEMS Yeast Res. 2017;17 doi: 10.1093/femsyr/fox059. [DOI] [PubMed] [Google Scholar]
  • 18.Shih JW, Wang WT, Tsai TY, Kuo CY, Li HK, Wu Lee YH. Critical roles of RNA helicase DDX3 and its interactions with eIF4E/PABP1 in stress granule assembly and stress response. Biochem J. 2012;441:119–29. doi: 10.1042/BJ20110739. [DOI] [PubMed] [Google Scholar]
  • 19.Shelkovnikova TA, Dimasi P, Kukharsky MS, An H, Quintiero A, Schirmer C, Buée L, Galas MC, Buchman VL. Chronically stressed or stress-preconditioned neurons fail to maintain stress granule assembly. Cell Death Dis. 2017;8:e2788. doi: 10.1038/cddis.2017.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Donnelly N, Gorman AM, Gupta S, Samali A. The eIF2α kinases: their structures and functions. Cell Mol Life Sci. 2013;70:3493–511. doi: 10.1007/s00018-012-1252-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Taniuchi S, Miyake M, Tsugawa K, Oyadomari M, Oyadomari S. Integrated stress response of vertebrates is regulated by four eIF2α kinases. Sci Rep. 2016;6:32886. doi: 10.1038/srep32886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Panas MD, Ivanov P, Anderson P. Mechanistic insights into mammalian stress granule dynamics. J Cell Biol. 2016;215:313–323. doi: 10.1083/jcb.201609081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chesnokova E, Bal N, Kolosov P. Kinases of eIF2a switch translation of mrna subset during neuronal plasticity. Int J Mol Sci. 2017;18:2213. doi: 10.3390/ijms18102213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhang S, Macias-Garcia A, Ulirsch JC, Velazquez J, Butty VL, Levine SS, Sankaran VG, Chen JJ. HRI coordinates translation necessary for protein homeostasis and mitochondrial function in erythropoiesis. Elife. 2019;8:e46976. doi: 10.7554/eLife.46976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fritzlar S, Aktepe TE, Chao YW, Kenney ND, McAllaster MR, Wilen CB, White PA, Mackenzie JM. Mouse Norovirus infection arrests host cell translation uncoupled from the stress granule-PKR-eIF2α axis. mBio. 2019;10:e00960–19. doi: 10.1128/mBio.00960-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sharma NR, Majerciak V, Kruhlak MJ, Zheng ZM. KSHV inhibits stress granule formation by viral ORF57 blocking PKR activation. PLoS Pathog. 2017;13:e1006677. doi: 10.1371/journal.ppat.1006677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pandey VK, Mathur A, Khan MF, Kakkar P. Activation of PERK-eIF2α-ATF4 pathway contributes to diabetic hepatotoxicity: Attenuation of ER stress by Morin. Cell Signal. 2019;59:41–52. doi: 10.1016/j.cellsig.2019.03.008. [DOI] [PubMed] [Google Scholar]
  • 28.Averous J, Lambert-Langlais S, Mesclon F, Carraro V, Parry L, Jousse C, Bruhat A, Maurin AC, Pierre P, Proud CG, Fafournoux P. GCN2 contributes to mTORC1 inhibition by leucine deprivation through an ATF4 independent mechanism. Sci Rep. 2016;6:27698. doi: 10.1038/srep27698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Qin Q, Carroll K, Hastings C, Miller CL. Mammalian orthoreovirus escape from host translational shutoff correlates with stress granule disruption and is independent of eIF2alpha phosphorylation and PKR. J Virol. 2011;85:8798–810. doi: 10.1128/JVI.01831-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ayuso MI, Martínez-Alonso E, Regidor I, Alcázar A. Stress granule induction after brain ischemia is independent of eukaryotic translation initiation factor (eIF) 2α phosphorylation and is correlated with a decrease in eIF4B and eIF4E proteins. J Biol Chem. 2016;291:27252–27264. doi: 10.1074/jbc.M116.738989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fujimura K, Sasaki AT, Anderson P. Selenite targets eIF4E-binding protein-1 to inhibit translation initiation and induce the assembly of non-canonical stress granules. Nucleic Acids Res. 2012;40:8099–110. doi: 10.1093/nar/gks566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mahboubi H, Stochaj U. Cytoplasmic stress granules: dynamic modulators of cell signaling and disease. Biochim Biophys Acta Mol Basis Dis. 2017;1863:884–895. doi: 10.1016/j.bbadis.2016.12.022. [DOI] [PubMed] [Google Scholar]
  • 33.Shi Q, Zhu Y, Ma J, Chang K, Ding D, Bai Y, Gao K, Zhang P, Mo R, Feng K, Zhao X, Zhang L, Sun H, Jiao D, Chen Y, Sun Y, Zhao SM, Huang H, Li Y, Ren S, Wang C. Prostate cancer-associated SPOP mutations enhance cancer cell survival and docetaxel resistance by upregulating Caprin1-dependent stress granule assembly. Mol Cancer. 2019;18:170. doi: 10.1186/s12943-019-1096-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wheeler JR, Matheny T, Jain S, Abrisch R, Parker R. Distinct stages in stress granule assembly and disassembly. Elife. 2016;5:e18413. doi: 10.7554/eLife.18413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364–78. doi: 10.1016/j.ejphar.2014.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yu W, Chen Y, Dubrulle J, Stossi F, Putluri V, Sreekumar A, Putluri N, Baluya D, Lai SY, Sandulache VC. Cisplatin generates oxidative stress which is accompanied by rapid shifts in central carbon metabolism. Sci Rep. 2018;8:4306. doi: 10.1038/s41598-018-22640-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pons DG, Nadal-Serrano M, Torrens-Mas M, Valle A, Oliver J, Roca P. UCP2 inhibition sensitizes breast cancer cells to therapeutic agents by increasing oxidative stress. Free Radic Biol Med. 2015;86:67–77. doi: 10.1016/j.freeradbiomed.2015.04.032. [DOI] [PubMed] [Google Scholar]
  • 38.Wu Z, Wang H, Fang S, Xu C. Roles of endoplasmic reticulum stress and autophagy on H2O2‑induced oxidative stress injury in HepG2 cells. Mol Med Rep. 2018;18:4163–4174. doi: 10.3892/mmr.2018.9443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kaehler C, Isensee J, Nonhoff U, Terrey M, Hucho T, Lehrach H, Krobitsch S. Ataxin-2-like is a regulator of stress granules and processing bodies. PLoS One. 2012;7:e50134. doi: 10.1371/journal.pone.0050134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lin L, Li X, Pan C, Lin W, Shao R, Liu Y, Zhang J, Luo Y, Qian K, Shi M, Bin J, Liao Y, Liao W. ATXN2L upregulated by epidermal growth factor promotes gastric cancer cell invasiveness and oxaliplatin resistance. Cell Death Dis. 2019;10:173. doi: 10.1038/s41419-019-1362-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, Zitvogel L, Kroemer G. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007;14:1848–50. doi: 10.1038/sj.cdd.4402201. [DOI] [PubMed] [Google Scholar]
  • 42.Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, Michaud M, Menger L, Gdoura A, Tajeddine N, Tesniere A, Zitvogel L, Kroemer G. Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress. Oncogene. 2011;30:1147–58. doi: 10.1038/onc.2010.500. [DOI] [PubMed] [Google Scholar]
  • 43.Vilas-Boas Fde A, da Silva AM, de Sousa LP, Lima KM, Vago JP, Bittencourt LF, Dantas AE, Gomes DA, Vilela MC, Teixeira MM, Barcelos LS. Impairment of stress granule assembly via inhibition of the eIF2alpha phosphorylation sensitizes glioma cells to chemotherapeutic agents. J Neurooncol. 2016;127:253–60. doi: 10.1007/s11060-015-2043-3. [DOI] [PubMed] [Google Scholar]
  • 44.Wang S, Kwon SH, Su Y, Dong Z. Stress granules are formed in renal proximal tubular cells during metabolic stress and ischemic injury for cell survival. Am J Physiol Renal Physiol. 2019;317:F116–F123. doi: 10.1152/ajprenal.00139.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rufo N, Garg AD, Agostinis P. The unfolded protein response in immunogenic cell death and cancer immunotherapy. Trends Cancer. 2017;3:643–658. doi: 10.1016/j.trecan.2017.07.002. [DOI] [PubMed] [Google Scholar]
  • 46.Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer. 2003;3:330–8. doi: 10.1038/nrc1074. [DOI] [PubMed] [Google Scholar]
  • 47.Mojardín L, Botet J, Quintales L, Moreno S, Salas M. New insights into the RNA-based mechanism of action of the anticancer drug 5’-fluorouracil in eukaryotic cells. PLoS One. 2013;8:e78172. doi: 10.1371/journal.pone.0078172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.García MA, Carrasco E, Aguilera M, Alvarez P, Rivas C, Campos JM, Prados JC, Calleja MA, Esteban M, Marchal JA, Aránega A. The chemotherapeutic drug 5-fluorouracil promotes PKR-mediated apoptosis in a p53-independent manner in colon and breast cancer cells. PLoS One. 2011;6:e23887. doi: 10.1371/journal.pone.0023887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ortega-García MB, Mesa A, Moya ELJ, Rueda B, Lopez-Ordoño G, García JÁ, Conde V, Redondo-Cerezo E, Lopez-Hidalgo JL, Jiménez G, Peran M, Martínez-González LJ, Del Val C, Zwir I, Marchal JA, García MÁ. Uncovering tumour heterogeneity through PKR and nc886 analysis in metastatic colon cancer patients treated with 5-FU-based chemotherapy. Cancers (Basel) 2020;12:379. doi: 10.3390/cancers12020379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kaehler C, Isensee J, Hucho T, Lehrach H, Krobitsch S. 5-Fluorouracil affects assembly of stress granules based on RNA incorporation. Nucleic Acids Res. 2014;42:6436–47. doi: 10.1093/nar/gku264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cheng ZF, Pai RK, Cartwright CA. Rack1 function in intestinal epithelia: regulating crypt cell proliferation and regeneration and promoting differentiation and apoptosis. Am J Physiol Gastrointest Liver Physiol. 2018;314:G1–G13. doi: 10.1152/ajpgi.00240.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Arimoto K, Fukuda H, Imajoh-Ohmi S, Saito H, Takekawa M. Formation of stress granules inhibits apoptosis by suppressing stress-responsive MAPK pathways. Nat Cell Biol. 2008;10:1324–32. doi: 10.1038/ncb1791. [DOI] [PubMed] [Google Scholar]
  • 53.Kwon HC, Moon CH, Kim SH, Choi HJ, Lee HS, Roh MS, Hwang TH, Kim JS, Kim HJ. Expression of double-stranded RNA-activated protein kinase (PKR) and its prognostic significance in lymph node negative rectal cancer. Jpn J Clin Oncol. 2005;35:545–50. doi: 10.1093/jjco/hyi146. [DOI] [PubMed] [Google Scholar]
  • 54.Xu C, Gamil A, Munang’andu HM, Evensen Ø. Apoptosis induction by dsRNA-dependent protein kinase R (PKR) in EPC cells via caspase 8 and 9 pathways. Viruses. 2018;10:526. doi: 10.3390/v10100526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Abaza Y, Kantarjian H, Garcia-Manero G, Estey E, Borthakur G, Jabbour E, Faderl S, O’Brien S, Wierda W, Pierce S, Brandt M, McCue D, Luthra R, Patel K, Kornblau S, Kadia T, Daver N, DiNardo C, Jain N, Verstovsek S, Ferrajoli A, Andreeff M, Konopleva M, Estrov Z, Foudray M, McCue D, Cortes J, Ravandi F. Long-term outcome of acute promyelocytic leukemia treated with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab. Blood. 2017;129:1275–1283. doi: 10.1182/blood-2016-09-736686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Valenzuela M, Glorieux C, Stockis J, Sid B, Sandoval JM, Felipe KB, Kviecinski MR, Verrax J, Buc Calderon P. Retinoic acid synergizes ATO-mediated cytotoxicity by precluding Nrf2 activity in AML cells. Br J Cancer. 2014;111:874–82. doi: 10.1038/bjc.2014.380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Iriyama N, Yuan B, Hatta Y, Horikoshi A, Yoshino Y, Toyoda H, Aizawa S, Takeuchi J. Granulocyte colony-stimulating factor potentiates differentiation induction by all-trans retinoic acid and arsenic trioxide and enhances arsenic uptake in the acute promyelocytic leukemia cell line HT93A. Oncol Rep. 2012;28:1875–82. doi: 10.3892/or.2012.2006. [DOI] [PubMed] [Google Scholar]
  • 58.Sun Y, Wang C, Wang L, Dai Z, Yang K. Arsenic trioxide induces apoptosis and the formation of reactive oxygen species in rat glioma cells. Cell Mol Biol Lett. 2018;23:13. doi: 10.1186/s11658-018-0074-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Martelli MP, Gionfriddo I, Mezzasoma F, Milano F, Pierangeli S, Mulas F, Pacini R, Tabarrini A, Pettirossi V, Rossi R, Vetro C, Brunetti L, Sportoletti P, Tiacci E, Di Raimondo F, Falini B. Arsenic trioxide and all-trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1-mutated AML cells. Blood. 2015;125:3455–65. doi: 10.1182/blood-2014-11-611459. [DOI] [PubMed] [Google Scholar]
  • 60.Wippich F, Bodenmiller B, Trajkovska MG, Wanka S, Aebersold R, Pelkmans L. Dual specificity kinase DYRK3 couples stress granule condensation/dissolution to mTORC1 signaling. Cell. 2013;152:791–805. doi: 10.1016/j.cell.2013.01.033. [DOI] [PubMed] [Google Scholar]
  • 61.Ozpolat B, Akar U, Tekedereli I, Alpay SN, Barria M, Gezgen B, Zhang N, Coombes K, Kornblau S, Lopez-Berestein G. PKCδ Regulates translation Initiation through PKR and eIF2α in response to retinoic acid in acute myeloid leukemia cells. Leuk Res Treatment. 2012;2012:482905. doi: 10.1155/2012/482905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Masciarelli S, Capuano E, Ottone T, Divona M, De Panfilis S, Banella C, Noguera NI, Picardi A, Fontemaggi G, Blandino G, Lo-Coco F, Fazi F. Retinoic acid and arsenic trioxide sensitize acute promyelocytic leukemia cells to ER stress. Leukemia. 2018;32:285–294. doi: 10.1038/leu.2017.231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Srivastava RK, Li C, Ahmad A, Abrams O, Gorbatyuk MS, Harrod KS, Wek RC, Afaq F, Athar M. ATF4 regulates arsenic trioxide-mediated NADPH oxidase, ER-mitochondrial crosstalk and apoptosis. Arch Biochem Biophys. 2016;609:39–50. doi: 10.1016/j.abb.2016.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen PX, Li QY, Yang Z. Musashi-1 expression is a prognostic factor in ovarian adenocarcinoma and correlates with ALDH-1 expression. Pathol Oncol Res. 2015;21:1133–40. doi: 10.1007/s12253-015-9943-6. [DOI] [PubMed] [Google Scholar]
  • 65.Nikpour P, Baygi ME, Steinhoff C, Hader C, Luca AC, Mowla SJ, Schulz WA. The RNA binding protein Musashi1 regulates apoptosis, gene expression and stress granule formation in urothelial carcinoma cells. J Cell Mol Med. 2011;15:1210–24. doi: 10.1111/j.1582-4934.2010.01090.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chen HY, Lin LT, Wang ML, Tsai KL, Huang PI, Yang YP, Lee YY, Chen YW, Lo WL, Lan YT, Chiou SH, Lin CM, Ma HI, Chen MT. Musashi-1 promotes chemoresistant granule formation by PKR/eIF2α signalling cascade in refractory glioblastoma. Biochim Biophys Acta Mol Basis Dis. 2018;1864:1850–1861. doi: 10.1016/j.bbadis.2018.02.017. [DOI] [PubMed] [Google Scholar]
  • 67.Glass L, Wente SR. Gle1 mediates stress granule-dependent survival during chemotoxic stress. Adv Biol Regul. 2019;71:156–171. doi: 10.1016/j.jbior.2018.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Steinmetz MO, Prota AE. Microtubule-targeting agents: strategies to hijack the cytoskeleton. Trends Cell Biol. 2018;28:776–792. doi: 10.1016/j.tcb.2018.05.001. [DOI] [PubMed] [Google Scholar]
  • 69.Bates D, Eastman A. Microtubule destabilising agents: far more than just antimitotic anticancer drugs. Br J Clin Pharmacol. 2017;83:255–268. doi: 10.1111/bcp.13126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chernov KG, Barbet A, Hamon L, Ovchinnikov LP, Curmi PA, Pastré D. Role of microtubules in stress granule assembly: microtubule dynamical instability favors the formation of micrometric stress granules in cells. J Biol Chem. 2009;284:36569–80. doi: 10.1074/jbc.M109.042879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ivanov PA, Chudinova EM, Nadezhdina ES. Disruption of microtubules inhibits cytoplasmic ribonucleoprotein stress granule formation. Exp Cell Res. 2003;290:227–33. doi: 10.1016/s0014-4827(03)00290-8. [DOI] [PubMed] [Google Scholar]
  • 72.Szaflarski W, Fay MM, Kedersha N, Zabel M, Anderson P, Ivanov P. Vinca alkaloid drugs promote stress-induced translational repression and stress granule formation. Oncotarget. 2016;7:30307–22. doi: 10.18632/oncotarget.8728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Piñeiro D, González VM, Hernández-Jiménez M, Salinas M, Martín ME. Translation regulation after taxol treatment in NIH3T3 cells involves the elongation factor (eEF)2. Exp Cell Res. 2007;313:3694–706. doi: 10.1016/j.yexcr.2007.07.025. [DOI] [PubMed] [Google Scholar]
  • 74.Palumbo A, Chanan-Khan A, Weisel K, Nooka AK, Masszi T, Beksac M, Spicka I, Hungria V, Munder M, Mateos MV, Mark TM, Qi M, Schecter J, Amin H, Qin X, Deraedt W, Ahmadi T, Spencer A, Sonneveld P CASTOR Investigators. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med. 2016;375:754–66. doi: 10.1056/NEJMoa1606038. [DOI] [PubMed] [Google Scholar]
  • 75.Chen RW, Palmer JM, Tomassetti S, Popplewell LL, Alluin J, Chomchan P, Nademanee AP, Siddiqi T, Tsai NC, Chen L, Zuo F, Abary R, Cai JL, Herrera AF, Rossi JJ, Rosen ST, Forman SJ, Kwak LW, Holmberg LA. Multi-center phase II trial of bortezomib and rituximab maintenance combination therapy in patients with mantle cell lymphoma after consolidative autologous stem cell transplantation. J Hematol Oncol. 2018;11:87. doi: 10.1186/s13045-018-0631-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cerruti F, Jocollè G, Salio C, Oliva L, Paglietti L, Alessandria B, Mioletti S, Donati G, Numico G, Cenci S, Cascio P. Proteasome stress sensitizes malignant pleural mesothelioma cells to bortezomib-induced apoptosis. Sci Rep. 2017;7:17626. doi: 10.1038/s41598-017-17977-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fournier MJ, Gareau C, Mazroui R. The chemotherapeutic agent bortezomib induces the formation of stress granules. Cancer Cell Int. 2010;10:12. doi: 10.1186/1475-2867-10-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Bittencourt L, Negreiros-Lima GL, Sousa LP, Silva AG, Souza IBS, Ribeiro RIMA, Dutra MF, Silva RF, Dias ACF, Soriani FM, Martins WK, Barcelos LS. G3BP1 knockdown sensitizes U87 glioblastoma cell line to Bortezomib by inhibiting stress granules assembly and potentializing apoptosis. J Neurooncol. 2019;144:463–473. doi: 10.1007/s11060-019-03252-6. [DOI] [PubMed] [Google Scholar]
  • 79.Gareau C, Fournier MJ, Filion C, Coudert L, Martel D, Labelle Y, Mazroui R. p21(WAF1/CIP1) upregulation through the stress granule-associated protein CUGBP1 confers resistance to bortezomib-mediated apoptosis. PLoS One. 2011;6:e20254. doi: 10.1371/journal.pone.0020254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Christen KE, Davis RA, Kennedy D. Psammaplysin F increases the efficacy of bortezomib and sorafenib through regulation of stress granule formation. Int J Biochem Cell Biol. 2019;112:24–38. doi: 10.1016/j.biocel.2019.04.008. [DOI] [PubMed] [Google Scholar]
  • 81.Zhang ZM, Wu JF, Luo QC, Liu QF, Wu QW, Ye GD, She HQ, Li BA. Pygo2 activates MDR1 expression and mediates chemoresistance in breast cancer via the Wnt/β-catenin pathway. Oncogene. 2016;35:4787–97. doi: 10.1038/onc.2016.10. [DOI] [PubMed] [Google Scholar]
  • 82.Henderson KA, Kobylewski SE, Yamada KE, Eckhert CD. Boric acid induces cytoplasmic stress granule formation, eIF2α phosphorylation, and ATF4 in prostate DU-145 cells. Biometals. 2015;28:133–41. doi: 10.1007/s10534-014-9809-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Unsworth H, Raguz S, Edwards HJ, Higgins CF, Yagüe E. mRNA escape from stress granule sequestration is dictated by localization to the endoplasmic reticulum. FASEB J. 2010;24:3370–80. doi: 10.1096/fj.09-151142. [DOI] [PubMed] [Google Scholar]
  • 84.Yagüe E, Raguz S. Escape from stress granule sequestration: another way to drug resistance. Biochem Soc Trans. 2010;38:1537–42. doi: 10.1042/BST0381537. [DOI] [PubMed] [Google Scholar]
  • 85.Flescher E, Rotem R. Protein kinase C epsilon mediates the induction of P-glycoprotein in LNCaP prostate carcinoma cells. Cell Signal. 2002;14:37–43. doi: 10.1016/s0898-6568(01)00215-7. [DOI] [PubMed] [Google Scholar]
  • 86.Kobayashi T, Winslow S, Sunesson L, Hellman U, Larsson C. PKCα binds G3BP2 and regulates stress granule formation following cellular stress. PLoS One. 2012;7:e35820. doi: 10.1371/journal.pone.0035820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Zhan Y, Fan S. Multiple mechanisms involving in radioresistance of nasopharyngeal carcinoma. J Cancer. 2020;11:4193–4204. doi: 10.7150/jca.39354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tsoumas D, Nikou S, Giannopoulou E, Champeris Tsaniras S, Sirinian C, Maroulis I, Taraviras S, Zolota V, Kalofonos HP, Bravou V. ILK Expression in colorectal cancer is associated with EMT, cancer stem cell markers and chemoresistance. Cancer Genomics Proteomics. 2018;15:127–141. doi: 10.21873/cgp.20071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wang Y, Fu D, Chen Y, Su J, Wang Y, Li X, Zhai W, Niu Y, Yue D, Geng H. G3BP1 promotes tumor progression and metastasis through IL-6/G3BP1/STAT3 signaling axis in renal cell carcinomas. Cell Death Dis. 2018;9:501. doi: 10.1038/s41419-018-0504-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gupta N, Badeaux M, Liu Y, Naxerova K, Sgroi D, Munn LL, Jain RK, Garkavtsev I. Stress granule-associated protein G3BP2 regulates breast tumor initiation. Proc Natl Acad Sci U S A. 2017;114:1033–1038. doi: 10.1073/pnas.1525387114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chiou GY, Yang TW, Huang CC, Tang CY, Yen JY, Tsai MC, Chen HY, Fadhilah N, Lin CC, Jong YJ. Musashi-1 promotes a cancer stem cell lineage and chemoresistance in colorectal cancer cells. Sci Rep. 2017;7:2172. doi: 10.1038/s41598-017-02057-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Shou Z, Jin X, He X, Zhao Z, Chen Y, Ye M, Yao J. Overexpression of Musashi-1 protein is associated with progression and poor prognosis of gastric cancer. Oncol Lett. 2017;13:3556–3566. doi: 10.3892/ol.2017.5879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ma L, Xu YL, Ding WJ, Shao HF, Teng YC. Prognostic value of Musashi-1 in endometrioid adenocarcinoma. Int J Clin Exp Pathol. 2015;8:4564–72. [PMC free article] [PubMed] [Google Scholar]
  • 94.Kudinov AE, Karanicolas J, Golemis EA, Boumber Y. Musashi RNA-binding proteins as cancer drivers and novel therapeutic targets. Clin Cancer Res. 2017;23:2143–2153. doi: 10.1158/1078-0432.CCR-16-2728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chen H, Liu J, Wang H, Cheng Q, Zhou C, Chen X, Ye F. Inhibition of RNA-binding protein musashi-1 suppresses malignant properties and reverses paclitaxel resistance in ovarian carcinoma. J Cancer. 2019;10:1580–1592. doi: 10.7150/jca.27352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Timalsina S, Arimoto-Matsuzaki K, Kitamura M, Xu X, Wenzhe Q, Ishigami-Yuasa M, Kagechika H, Hata Y. Chemical compounds that suppress hypoxia-induced stress granule formation enhance cancer drug sensitivity of human cervical cancer HeLa cells. J Biochem. 2018;164:381–391. doi: 10.1093/jb/mvy062. [DOI] [PubMed] [Google Scholar]
  • 97.Taylor MA, Das BC, Ray SK. Targeting autophagy for combating chemoresistance and radioresistance in glioblastoma. Apoptosis. 2018;23:563–575. doi: 10.1007/s10495-018-1480-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Krisenko MO, Higgins RL, Ghosh S, Zhou Q, Trybula JS, Wang WH, Geahlen RL. Syk is recruited to stress granules and promotes their clearance through autophagy. J Biol Chem. 2015;290:27803–15. doi: 10.1074/jbc.M115.642900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chang YJ, Li LT, Chen HA, Hung CS, Wei PL. Silencing survivin activates autophagy as an alternative survival pathway in HCC cells. Tumour Biol. 2014;35:9957–66. doi: 10.1007/s13277-014-2257-6. [DOI] [PubMed] [Google Scholar]
  • 100.Gao P, Qiao X, Sun H, Huang Y, Lin J, Li L, Wang X, Li C. Activated spleen tyrosine kinase promotes malignant progression of oral squamous cell carcinoma via mTOR/S6 signaling pathway in an ERK1/2-independent manner. Oncotarget. 2017;8:83900–83912. doi: 10.18632/oncotarget.19911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sfakianos AP, Mellor LE, Pang YF, Kritsiligkou P, Needs H, Abou-Hamdan H, Désaubry L, Poulin GB, Ashe MP, Whitmarsh AJ. The mTOR-S6 kinase pathway promotes stress granule assembly. Cell Death Differ. 2018;25:1766–1780. doi: 10.1038/s41418-018-0076-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lastres-Becker I, Nonis D, Eich F, Klinkenberg M, Gorospe M, Kötter P, Klein FA, Kedersha N, Auburger G. Mammalian ataxin-2 modulates translation control at the pre-initiation complex via PI3K/mTOR and is induced by starvation. Biochim Biophys Acta. 2016;1862:1558–69. doi: 10.1016/j.bbadis.2016.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Heberle AM, Prentzell MT, van Eunen K, Bakker BM, Grellscheid SN, Thedieck K. Molecular mechanisms of mTOR regulation by stress. Mol Cell Oncol. 2015;2:e970489. doi: 10.4161/23723548.2014.970489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Thedieck K, Holzwarth B, Prentzell MT, Boehlke C, Kläsener K, Ruf S, Sonntag AG, Maerz L, Grellscheid SN, Kremmer E, Nitschke R, Kuehn EW, Jonker JW, Groen AK, Reth M, Hall MN, Baumeister R. Inhibition of mTORC1 by astrin and stress granules prevents apoptosis in cancer cells. Cell. 2013;154:859–74. doi: 10.1016/j.cell.2013.07.031. [DOI] [PubMed] [Google Scholar]
  • 105.Zheng H, Zhan Y, Zhang Y, Liu S, Lu J, Yang Y, Wen Q, Fan S. Elevated expression of G3BP1 associates with YB1 and p-AKT and predicts poor prognosis in nonsmall cell lung cancer patients after surgical resection. Cancer Med. 2019;8:6894–6903. doi: 10.1002/cam4.2579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hofmann S, Cherkasova V, Bankhead P, Bukau B, Stoecklin G. Translation suppression promotes stress granule formation and cell survival in response to cold shock. Mol Biol Cell. 2012;23:3786–800. doi: 10.1091/mbc.E12-04-0296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Reineke LC, Cheema SA, Dubrulle J, Neilson JR. Chronic starvation induces noncanonical pro-death stress granules. J Cell Sci. 2018;131:jcs220244. doi: 10.1242/jcs.220244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mazan-Mamczarz K, Peroutka RJ, Steinhardt JJ, Gidoni M, Zhang YQ, Lehrmann E, Landon AL, Dai BJ, Houng S, Muniandy PA, Efroni S, Becker KG, Gartenhaus RB. Distinct inhibitory effects on mTOR signaling by ethanol and INK128 in diffuse large B-cell lymphoma. Cell Commun Signal. 2015;13:15. doi: 10.1186/s12964-015-0091-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Heberle AM, Razquin Navas P, Langelaar-Makkinje M, Kasack K, Sadik A, Faessler E, Hahn U, Marx-Stoelting P, Opitz CA, Sers C, Heiland I, Schäuble S, Thedieck K. The PI3K and MAPK/p38 pathways control stress granule assembly in a hierarchical manner. Life Sci Alliance. 2019;2:e201800257. doi: 10.26508/lsa.201800257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Salaroglio IC, Panada E, Moiso E, Buondonno I, Provero P, Rubinstein M, Kopecka J, Riganti C. PERK induces resistance to cell death elicited by endoplasmic reticulum stress and chemotherapy. Mol Cancer. 2017;16:91. doi: 10.1186/s12943-017-0657-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Grabocka E, Bar-Sagi D. Mutant KRAS enhances tumor cell fitness by upregulating stress granules. Cell. 2016;167:1803–1813.e12. doi: 10.1016/j.cell.2016.11.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chen ZH, Qi JJ, Wu QN, Lu JH, Liu ZX, Wang Y, Hu PS, Li T, Lin JF, Wu XY, Miao L, Zeng ZL, Xie D, Ju HQ, Xu RH, Wang F. Eukaryotic initiation factor 4A2 promotes experimental metastasis and oxaliplatin resistance in colorectal cancer. J Exp Clin Cancer Res. 2019;38:196. doi: 10.1186/s13046-019-1178-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Xi C, Wang L, Yu J, Ye H, Cao L, Gong Z. Inhibition of eukaryotic translation initiation factor 4E is effective against chemo-resistance in colon and cervical cancer. Biochem Biophys Res Commun. 2018;503:2286–2292. doi: 10.1016/j.bbrc.2018.06.150. [DOI] [PubMed] [Google Scholar]
  • 114.Martínez A, Sesé M, Losa JH, Robichaud N, Sonenberg N, Aasen T, Ramón Y Cajal S. Phosphorylation of eIF4E confers resistance to cellular stress and DNA-damaging agents through an interaction with 4E-T: a rationale for novel therapeutic approaches. PLoS One. 2015;10:e0123352. doi: 10.1371/journal.pone.0123352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bell JB, Eckerdt F, Dhruv HD, Finlay D, Peng S, Kim S, Kroczynska B, Beauchamp EM, Alley K, Clymer J, Goldman S, Cheng SY, James CD, Nakano I, Horbinski C, Mazar AP, Vuori K, Kumthekar P, Raizer J, Berens ME, Platanias LC. Differential response of glioma stem cells to arsenic trioxide therapy is regulated by MNK1 and mRNA translation. Mol Cancer Res. 2018;16:32–46. doi: 10.1158/1541-7786.MCR-17-0397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sun Y, Jiang Y, Huang J, Chen H, Liao Y, Yang Z. CISD2 enhances the chemosensitivity of gastric cancer through the enhancement of 5-FU-induced apoptosis and the inhibition of autophagy by AKT/mTOR pathway. Cancer Med. 2017;6:2331–2346. doi: 10.1002/cam4.1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Milošević Z, Banković J, Dinić J, Tsimplouli C, Sereti E, Dragoj M, Paunović V, Milovanović Z, Stepanović M, Tanić N, Dimas K, Pešić M. Potential of the dual mTOR kinase inhibitor AZD2014 to overcome paclitaxel resistance in anaplastic thyroid carcinoma. Cell Oncol (Dordr) 2018;41:409–426. doi: 10.1007/s13402-018-0380-x. [DOI] [PubMed] [Google Scholar]
  • 118.Moeller BJ, Cao Y, Li CY, Dewhirst MW. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5:429–41. doi: 10.1016/s1535-6108(04)00115-1. [DOI] [PubMed] [Google Scholar]
  • 119.Suresh PS, Tsutsumi R, Venkatesh T. YBX1 at the crossroads of non-coding transcriptome, exosomal, and cytoplasmic granular signaling. Eur J Cell Biol. 2018;97:163–167. doi: 10.1016/j.ejcb.2018.02.003. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES