ABSTRACT
From initiation through progression, cancer cells are subjected to a magnitude of endogenous and exogenous stresses, which aid in their neoplastic transformation. Exposure to these classes of stress induces imbalance in cellular homeostasis and, in response, cancer cells employ informative adaptive mechanisms to rebalance biochemical processes that facilitate survival and maintain their existence. Different kinds of stress stimuli trigger epigenetic alterations in cancer cells, which leads to changes in their transcriptome and metabolome, ultimately resulting in suppression of growth inhibition or induction of apoptosis. Whether cancer cells show a protective response to stress or succumb to cell death depends on the type of stress and duration of exposure. A thorough understanding of epigenetic and molecular architecture of cancer cell stress response pathways can unveil a plethora of information required to develop novel anticancer therapeutics. The present view highlights current knowledge about alterations in epigenome and transcriptome of cancer cells as a consequence of exposure to different physicochemical stressful stimuli such as reactive oxygen species (ROS), hypoxia, radiation, hyperthermia, genotoxic agents, and nutrient deprivation. Currently, an anticancer treatment scenario involving the imposition of stress to target cancer cells is gaining traction to augment or even replace conventional therapeutic regimens. Therefore, a comprehensive understanding of stress response pathways is crucial for devising and implementing novel therapeutic strategies.
KEYWORDS: epigenetic response, oxidative stress, DNA damage, hypoxia, hyperthermia, nutrient deprivation
INTRODUCTION
One of the most critical aspects of cellular physiology is the maintenance of homeostasis, which is governed by evolutionarily conserved and interwoven molecular cascades. The extracellular milieu or microenvironment of cancer cells has a major impact on their internal molecular architecture. Throughout the course of initiation, progression, invasion, and metastasis, cancer cells constantly adapt to various forms of exogenous and endogenous stress stimuli that challenge their internal homeostatic balance. Changes in the microenvironment, such as altered oxygen and nutrient availability, largely impact the internal molecular and epigenetic landscape of the cancer cells. Likewise, internal imbalances in the molecular equilibrium caused by genomic instability, the accumulation of incorrectly folded proteins, oxidative stress, etc., lead to many alternations in cellular signaling pathways, which cumulatively determine cell fate. Whether the cancer cell will adapt to and survive the stressful stimuli or undergo apoptosis is guided by intricate signals and is largely determined by the nature and duration of the stress stimulus.
In this context, an understanding of the epigenetic alterations employed by cancer cells as an approach to adapt to stress is of crucial significance and can be utilized to devise a potential strategy to ameliorate anticancer treatment. Chemotherapy, the first-line therapeutic regimen utilized for most cancers, induces genotoxic stress to cancer cells. However, in the majority of cases the development of chemoresistance leads to tumor recurrence, which ultimately results in a poor prognosis. Several studies have indicated that cancer cells employ elaborate epigenetic cascades to adapt to chemotherapeutic and other stresses to develop therapeutic resistance (1, 2). A complete understanding of the stress-adapting epigenetic pathways of cancer cells can unveil a plethora of target molecules, which can in turn be modulated to attenuate the stress adaptations of cancer cells.
In this review, we present a comprehensive view of the current understanding on the alternations in epigenome and transcriptome levels in cancer cells when subjected to various physicochemical stress stimuli, such as oxidative stress, hypoxia, hyperthermia, alkylation, and radiation-mediated DNA damage, nutrient deficiency, and misfolded protein accumulation. This accrued information can enlighten some previously unexplored targets that can be potentially utilized to device novel therapeutic regimens against cancer.
KEY STRESS RESPONSE PATHWAYS AND THEIR EPIGENETIC AND MOLECULAR SIGNATURES
Oxidative stress and its epigenetic response.
Basal levels of reactive oxygen species (ROS) are constantly generated in each and every cell type, as a byproduct of metabolic pathways (3, 4). However, the cellular antioxidant mechanisms scavenge the ROS to diminish the ROS-mediated intracellular damage. For example, most ROS radicals react with oxygen to form superoxide, so the human cellular and mitochondrial superoxide dismutases are master regulators of ROS that use oxidation and reduction to dismutase superoxide to water and hydrogen peroxide, the latter being removed by catalase. Under various conditions, the subtle balance between ROS generation and ROS scavenging is perturbed, leading to the accumulation of elevated levels of ROS, thereby disrupting cellular homeostasis (5). As an example of how ROS can reset the cell to a higher level of oxidative stress, the cytoplasmic superoxide dismutase dimer has free cysteines, and their oxidation reduces the protein stability, thereby reducing ROS production and removal. Various conditions, such as hypoxia, nutrient imbalance, hyperthermia, and exposure to genotoxic agents, can lead to significantly augmented production of ROS within the cell, which ultimately leads to a state of oxidative stress. Nitric oxide synthesis, which is controlled by nitric oxide synthases, can also reduce oxidative stress in cancer cells by forming dinitrosyl complexes. Loss of ROS balance can alter many aspects of cell metabolism, including signaling and cell death.
(i) Altered epigenetic landscape in oxidative stress.
Heightened accumulation of intracellular ROS damages the biomolecules and disrupts the balance of cellular metabolites, thus perturbing genomic stability and compromising the intracellular epigenetic balance (Fig. 1, upper left panel). As a consequence of these alterations, senescence, cell death, and even cancer may ensue. For instance, S-adenosylmethionine (SAM) is used as a substrate by histone and DNA methyltransferases (6); ROS-mediated oxidization of GSH to GSSH attenuates SAM production by inhibiting the formation of SAM synthetase, the enzyme responsible for SAM production, thus disrupting methyltransferase activity (7, 8), leading to alterations in DNA and histone methylation. The role of ROS in mediating DNA damage and the cellular DNA damage response pathways in this context are well documented by numerous studies (9–11). It has been indicated that induction of oxidative stress recruits a DNMT1/DNMT3b/PRC4 complex to CpG islands on various gene promoters, causing hypermethylation and silencing of those genes (12).
FIG 1.
Schematic diagram representing the different epigenetic and molecular cascades employed by cancer cells to adapt in response to oxidative stress, hypoxia, hyperthermia, radiation, protein misfolding, and nutrient deprivation.
(ii) Oxidative stress and its effect on the epigenetic machinery in the context of cancer.
ROS-mediated DNA damage has been widely correlated with the development and progression of cancer (13). Intriguingly, based upon the evolutionarily conserved unstructured domain that targets the oxidized base excision repair enzyme NEIL1 to open chromatin, its damage surveillance of oxidation-susceptible sites to preserve essential gene function and to limit instability and cancer likely originated ∼500 million years ago during the build-up of free atmospheric oxygen. Indeed, the accumulation of ROS and oxidative stress is considered to be one of the prime reasons behind DNA damage and genomic instability, which ultimately leads to malignant transformation (14). ROS is reported to modulate the activities of various epigenomic modifiers, such as DNMT1, DNMT3a, histone deacetylase 1 (HDAC1) HMT1, HAT1, and MBD4, thus acting as a neoplastic trigger (15).
The role of oxidative stress in initiation of carcinogenesis has been studied by various groups. A report by Nishida et al. (16) on chronic hepatitis C indicates that 8-hydroxy-2′-deoxyguanosine, a ROS-mediated promutagenic DNA lesion, leads transformation of chromatin activation marks (H3K4me3 and H4K16ac) to a chromatin repression mark (H3K27me3) in the promoters of tumor suppressor genes. This, in turn, acts as a risk factor for the onset of hepatocellular carcinoma. A relatively older breast cancer study established the involvement of 8-hydroxy-2′-deoxyguanosine in the development of breast carcinoma (17). Another indicator of ROS-mediated DNA damage, 8-oxo-7,8-dihydro-2-deoxyguanosine, is reported to negatively correlate with the epigenetic marker 5-methylcytosine (m5C) and act as a prerequisite for development and progression of glioma (18).
The ingenious ability of cancer cells to utilize the heightened level of ROS to promote their survival is supported by various studies. For example, ROS-mediated hyperactivation of mitogen-activated protein kinase (19), NF-κB (20), peroxisome proliferator-activated receptor gamma (PPARγ) (21), and STAT3 (22) pathways stimulates cell proliferation and ensures the progression of cancer. A study by Lim et al. (23) on hepatocellular carcinoma indicated that ROS induces hypermethylation of the E-Cadherin promoter via recruitment of HDAC1 and DNMT1 by SNAIL, thus leading to activation of the epithelial-mesenchymal transition (EMT) and metastasis. A similar study on colorectal cancer indicated that ROS increases the binding of DNMT1 and HDAC1 to the tumor suppressor Runt domain transcription factor 3 (RUNX3) promoter, leading to its hypermethylation and suppression (24), thereby promoting colorectal cancer progression (24). Long interspersed nuclear element 1 (LINE-1) is the most abundant type of transposable element found in eukaryotic genomes. In normal somatic cells, LINE-1 elements are mostly methylated and suppressed. However, hypomethylated and active, LINE-1 elements lead to genomic instability and mutagenesis (25, 26). In another study in bladder cancer, ROS-mediated hypomethylation of LINE-1 was associated with heightened genomic instability and progression of cancer (27). Therefore, all these observations point toward the ingenious adaptive ability of cancer cells to oxidative stress that ultimately leads to heightened tumor progression and poorer prognostic outcome.
Epigenetic responses to hypoxia.
Oxygen is one of the most critical components supporting life of complex organisms, and in the human body it is a defining criterion of the metabolic landscape. Extremely scarce availability of oxygen in the cellular microenvironment (i.e., hypoxia) can potentially promote stress, altering the homeostasis of cells and their metabolic architecture. Hypoxia is a hallmark of the malignant phenotype and a key feature of the microenvironment in solid tumors, where the supply of oxygen is frequently irregular despite angiogenesis. The irregularity in the spatial distribution of vasculature leads to the onset of a steep oxygen gradient toward the tumor core, resulting in hypoxia and profoundly affecting the biological behavior of the hypoxic cancer cells. Hypoxia is reported to be associated with reduced sensitivity to radiotherapy or chemotherapy (28, 29) and an increased invasive and metastatic phenotype (30). Hypoxia induces both proteomic and genomic, as well as epigenomic, changes within tumor cells. In cancer cells, the response to hypoxia is governed by a heterodimeric transcriptional regulator termed hypoxia-inducible factor (HIF) (31, 32). In response to hypoxia, HIF-1α becomes stabilized by binding to a constitutively expressed β-subunit (HIF-1β), localizing to the nucleus and activating hypoxia response elements (HREs) to augment the expression of target genes (31, 33). In turn, HIF-1-activated genes regulate cellular glucose and oxygen consumption and glucose metabolic pathways. Several studies have indicated that HIF-1 induces the expression of stemness factors (34, 35) and promotes angiogenesis and metastasis in response to hypoxia (36–38).
(i) Epigenetic regulation of HIF-1α stability.
Stabilization of HIF-1α is one of the most crucial aspects in hypoxia adaptation. Under normal conditions, HIF-1α is hydroxylated by prolyl hydroxylases (PHD). Upon hydroxylation, its oxygen-dependent degradation (ODD) domain recognized by von Hippel-Lindau tumor suppressor protein (VHL), eventually leading to proteasomal degradation by the cullin2 E3 ligase complex (39–42). In contrast, under hypoxic stress, the enzymatic activity of PHDs decreases, thereby inhibiting the hydroxylation of HIF-1α while favoring its stabilization. In addition to hydroxylation, other posttranslational modifications, such as acetylation, phosphorylation, and SUMOylation, have been reported to modulate HIF-1α. Previous reports indicated that SENP1 de-SUMOylates and stabilizes HIF-1α (43); moreover, phosphorylation of HIF-1α by p38 inhibits VHL binding, leading to its stabilization (44). In contrast, HIF-1α acetylation is associated with VHL-mediated ubiquitination and degradation (45). A recent report has revealed the presence of a methylation/demethylation cycle involving methylation of HIF-1α by SET7/9 and demethylation by LSD1 as a way to regulate HIF-1α stability under hypoxic conditions (46). The epigenetic state of crucial members of HIF-1 signaling cascade also regulates HIF stability and influences its target gene expression. For instance, hypermethylation of the VHL promoter and its inactivation increases the stabilization of HIF-1α and promotes the expression of its target genes, such as carbonic anhydrase 9 (CA9) and glucose transporter type 1 (GLUT1) (47).
DNA and histone demethylation are also related to the dynamic regulation of HIF-1α. Indeed, it has been reported that in colon cancer, demethylation of CpG islands in the HIF-1α promoter promotes binding of HIF-1α to its own promoter, thus establishing a positive-feedback loop (48). Induction of euchromatic histone-lysine N-methyltransferase 2 (G9a) by hypoxic stimuli acts as a central crossroad in hypoxia-mediated signaling pathways (49, 50). G9a leads to enhancement of H3K9me2 and subsequent repression of several hypoxia-regulated genes (50, 51) (Fig. 1, upper middle panel). For instance, Reptin, a nonhistone chromatin modifier that modulates about 24.6% of hypoxia-responsive genes, is methylated and repressed by G9a (52). It binds to, deacetylates, and transcriptionally represses VEGF, BNIP3, and PGK1 via recruitment of HDAC1 (52). In contrast, G9a methylates Pontin, a chromatin-remodeling factor that regulates about 23.5% of hypoxia-responsive genes (53). Increased expression of these genes via methylated Pontin promotes hypoxia-induced tumor progression, EMT, and metastasis. Furthermore, G9a stimulates HIF-1α-mediated target gene activation by recruiting p300 to the promoters of HIF-1α target genes, including Ets1, KDM4B, and IGFBP3 (53). In addition, acetylase activity by p300/CBP and TAT-interactive protein 60 (TIP60) regulates the transcriptional expression of HIF-1α (54), while class I histone deacetylases (HDACs) regulate its stability (55). Under severe hypoxic stress, acetylation of HIF-1α at lysine 709 by p300 increases its stability (56). In contrast, SIRT1 (NAD-dependent deacetylase sirtuin 1) (Fig. 2) deacetylates p300/CBP-associated factor (PCAF)-mediated lysine acetylation of HIF-1α, thus preventing p300/CBP recruitment and hypoxia-induced target gene activation (57). SIRT3 has also been reported to inhibit HIF-1α, thereby suppressing the metabolic reprogramming of cancer cells and mitochondrial ROS production (58). Together, these reports highlight the epigenetic regulatory network of hypoxia-responsive gene expression through various histone and DNA alterations. The activation-inactivation cycle of HIF-1α is also regulated by protein acetylation: lysine 532 of HIF is acetylated by ARD1, which stimulates its interaction with VHL and subsequent degradation, under normoxic conditions (46). Under hypoxic stress, this state is reversed, supporting the activation of HIF-1α.
FIG 2.
SIRT1 is downregulated in cancer and associated with poor survival in patients with weak expression. (A) Box plot of SIRT1 mRNA levels in 15 tumor types from TCGA. Only tumors with at least 10 matched controls were included. (B) Kaplan-Meier survival curves and hazard ratios (HR) in patients with low (below mean) and high (above mean) SIRT1 mRNA levels in KIRC, LGG, and HNSC. Tumor abbreviations: BLCA, bladder urothelial carcinoma; BRCA, breast invasive carcinoma; CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma; COAD, colon adenocarcinoma; ESCA, esophageal carcinoma; HNSC, head and neck squamous cell carcinoma; KICH, kidney chromophobe; KIRC, kidney renal clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LGG, brain lower grade glioma; LIHC, liver hepatocellular carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; PRAD, prostate adenocarcinoma; STAD, stomach adenocarcinoma; THCA, thyroid carcinoma; THYM, thymoma; UCEC, uterine corpus endometrial carcinoma. Analyses were conducted as described previously (229).
(ii) Noncoding RNA-mediated regulation of HIF-1α.
The stability of HIF-1α in hypoxic cancer cells was also reported to be mediated by several noncoding RNAs. In hypoxic nasopharyngeal carcinoma cells, two long noncoding RNAs (lncRNAs), PVT1 and DANCR, play a role in stabilizing both HIF-1α mRNA and the protein, promoting hypoxia-induced tumor progression (59). Both PVT1 and DANCR are reported to be associated with poor prognosis of nasopharyngeal carcinoma (59, 60). MicroRNA 181c (miRNA-181c) in breast cancer (61) and miRNA-646 and lncRNA-MTA2TR in pancreatic cancer (62) are also involved in the regulation of HIF-1α stability and its accumulation, thus playing part in managing hypoxic stress in these cancer cells.
(iii) Epigenetic regulatory mechanisms behind hypoxia-induced EMT.
It is well established that hypoxia promotes tumor progression and metastasis. Recent reports have indicated that hypoxia-induced EMT is partly driven by HDAC3 and that its knockdown significantly impairs the metastatic potential of cancer cells under hypoxic stress (63, 64) (Fig. 1, upper middle panel). In hypoxic stress, HDAC3 is recruited to epithelial gene promoters, deacetylating H3K4 and repressing them (64). However, a bivalent domain of enhanced H3K4me2/me3 and H3K27me3 was recently reported on the promoters of epithelial genes (65). These bivalent domains supposedly maintain EMT genes repressed, while keeping them poised for activation at a later stage during mesenchymal-epithelial transition (MET). These observations support the activity of epigenetic modulatory mechanisms involved in hypoxia-induced alterations in tumor cells.
Epigenetic responses to heat stress. (i) Heat shock factors and heat shock proteins.
Hyperthermia, or heat stress, is defined as a temperature above physiologically optimum range which affects physiological and molecular properties (66). Hyperthermia activates a subset of ataxia-telangiectasia mutated effectors independent of DNA strand breaks (67). When a cell is exposed to heat stress, heat shock factors (HSFs), a family of transcription factors, regulate the expression of heat shock proteins (HSPs), an evolutionary conserved pathway known as heat shock response. The function of HSFs was first observed by the Italian geneticist Ferruccio Ritossa, who showed an induction of chromosome puffs in the salivary gland of the larvae of Drosophila busckii when exposed to an elevated temperature (68). Subsequent investigations indicated that these chromosomal puffs were the regions transcribing HSPs, which function as molecular chaperones (69). HSFs are responsible for mediating cellular adaptations to proteotoxic stress conditions, such as elevated temperature, pathogens, toxins, etc. (70, 71). It has also been suggested that HSFs play a critical role during differentiation processes such as neurogenesis and gametogenesis (70, 71).
(ii) Role of heat shock factors in cancer.
HSFs and HSPs have been studied for decades in the context of cancer. HSPs have long been correlated with evasion of apoptosis, malignant transformation, and tumor progressions, and HSP inhibitors as an approach to target cancer cells are under investigation. More recent cancer research has focused on HSFs for their ability to transcriptionally regulate HSPs.
Mammalian HSFs are a family of five transcription factors and include HSF1, HSF2, HSF3, HSF4, and HSFγ. Among these, the functional regulation of HSF1, -2, and -4 has been mostly studied (72). HSF1 has been associated with tumor progression in several studies. For instance, HSF1-null mice are significantly resistant to dimethylbenzanthracene (DMBA)-induced skin cancer and mutant p53-induced malignant transformation (73). Studies have also reported resistance to HER2/ErbB2-induced breast cancer and carcinogen-induced liver cancer in humans and in HSF1-null mice (73–75). Overexpression of HSF1 is commonly observed in various carcinomas, and its higher expression is associated with poor prognosis (76). A meta-analysis of 10,287 cancer genomes from The Cancer Genome Atlas (TCGA) and cBioPortal databases revealed that the elevated expression of HSF1 in various types of cancers is due to the amplification of chromosome 8q24.3, where the HSF1 gene resides (77). In colorectal cancer, HSF1 reportedly promotes carcinogenesis by recruiting DNMT3a on MIR137HG. This leads to the suppression of miR-137, which in turn increases the expression of GLS1 and induces GLS1-mediated mTOR activation, thus promoting tumorigenesis (78). Inhibition or knockdown of HSF1 has been shown to reduce the tumor burden and to suppress growth in colorectal cancer cells (78).
HSF2 also plays a significant role in cancer. Once activated, HSF2 asserts its function by upregulating the expression of various HSPs, such as HSP27, HSP47, HSP70, and HSP90, thus promoting cell proliferation and migration (79). Along with HSF1 and HSF2, the role of HSF4 in cancer has been highlighted recently. Loss of HSF4 expression hinders Arf/p53 mediated tumorigenesis (80); however, unlike other HSFs, HSF4 also regulates cellular senescence (80) and, when inhibited, leads to cellular senescence and inhibition of cell proliferation. These roles for HSF4 depend upon the expression of p21 and p27 but are independent of p53 (80). In summary, a comprehensive understanding of the HSF family provides for selective optimization of various possibilities of new therapeutic interventions for cancer.
(iii) DNA damage and epigenetic alterations in hyperthermia.
Among the various cellular effects of hyperthermia, one of the key responses is the induction of DNA breaks and chromosomal aberrations. This response to hyperthermia occurs at temperatures higher than 41.5°C and involves focal phosphorylation of H2AX at serine 139 (γH2AX), as seen in response to double-strand breaks (81) (Fig. 1, upper right panel). Recent studies have indicated that at higher temperatures, i.e., 43 to 45.5°C, there is increased induction of γH2AX foci, a process which shows dependence on the DSB signaling factor ATM (67, 82, 83). In this context, various studies have reported a negative association between hyperthermia and the nonhomologous end-joining (NHEJ) DNA repair pathway (84, 85). A hyperthermia treatment at temperatures of 44 to 45°C inactivates DNA binding by KU70/80 and simultaneously decreases DNA-PK activity (86–89) and the levels of BRCA1 and 53BP1 (90). In general, we predict that DNA damage responses dependent upon conformational switching, and flexibility, such as the NHEJ repair complex, will be susceptible to hyperthermia treatments (91).
Apart from NHEJ, hyperthermia also inhibits repair by homologous recombination (HR) in both human and murine cells by inducing BRCA2 degradation (92). It also reduces the levels of MRE11 and inhibits the interaction with members of the Mre11-Rad51-Nbs1 (MRN) complex, which initiates HR, thereby abolishing HR-mediated DSB repair (93–96). Thus, there is a synergistic amplification of hyperthermia effects due to its impact upon depletion of key DNA damage response (DDR) proteins. That is, the structural biochemistry of DDR is connected to the cell biology of epigenetics and chromatin dynamics. For example, MRN is a primary sensor in the process of ATM activation and chromatin remodeling at DSBs and is required for establishment of epigenetic marks after repair DNA synthesis via the ATM kinase (97).
Inhibition of both of these repair pathways by hyperthermia depends on H4K16ac status. Specifically, depletion of hMOF, one of the histone acetyl transferases and a member of the largest lysine acetyltransferase (KAT) subdivision of the MYST family (MOZ, MOF, Ybf2, Sas2, and Tip60), plays a critical role (98) and leads to the attenuation of H4K16ac, which in turn causes a delay in the γH2AX focus formation (25). Therefore, regulation of H4K16ac by MOF directly impacts chromatin structure and thus affects the DDR pathway (25, 26). MOF was also reported to interact with several other proteins of the DDR pathway, regulating their functionality (25). All of these reports establish the significance of hMOF and hyperthermia in cancer therapeutics, in line with other studies reporting tumor regression following a moderate elevation in temperature (<45°C) (99).
Epigenetic alterations in radiation stress.
Ionizing radiation (IR) is the most extensively studied mutagenic radiating agent. Examples of IR include microwaves, X-rays, and gamma rays. This radiation can induce a vast array of DNA alterations, including nucleotide modifications, single- and double-strand breaks, and the formation of DNA-DNA and DNA-protein cross-links. IR is also reported to cause chromosomal alterations, such as inversions and deletions, which are observed at metaphase. The extent of chromosomal aberrations caused by IR depends on the level of linear energy transfer (LET) (100), a measure of the density of ionization along the radiation beam. For instance, high-LET radiation can induce more complex chromosomal rearrangements than does low-LET radiation (101).
(i) IR-induced changes in DNA methylation pattern.
DNA methylation is one of the most common epigenetic alterations. Accumulating evidence indicates that variations in DNA methylation patterns might facilitate the sensitivity or resistance of tumor cells to IR. Kalinich et al. (102) indicated that cancer cells, when exposed to 60Co gamma irradiation in vitro, show a dose-dependent decrease in the level of 5-methylcytosine. Further studies confirmed that low-LET radiation induces genome-wide DNA hypomethylation.
Radiation exposure alters DNA methylation in a locus-specific manner. A study in triple-negative breast cancer by Antwih et al. (103) reported that RB1, IGF1R, and KRAS genes were differentially methylated after radiation exposure and that their levels of transcriptional expression corresponded to the level of methylation. This study also revealed altered expression of DNMT1 following X-ray radiation above 6 Gy (103). Various in vivo studies have indicated that X-rays can induce loss of global DNA methylation in target-specific and -nonspecific tissues (104–106). The alternations in methylation profile following radiation exposure have been studied in murine models also. Koturbash et al. (107) reported that acute and fractionated whole-body irradiation of mice leads to significant decreases in cytosine DNA methylation in the thymus, which in turn predisposes mice to lymphoma. In addition, irradiation-induced murine lymphomas showed an increased frequency of homozygous deletions of the Rit1/Bcl11b tumor-suppressor genes (107). The same group reported that a 1-Gy X-ray exposure for 6 h induced global DNA hypomethylation in murine spleen tissues (108).
LINE-1 hypomethylation has been observed in nearly all human cancers and is commonly associated with a poor prognosis. Several reports have indicated that exposure to IR leads to CpG hypomethylation and activation of LINE-1 retrotransposition activity in various tissues (109–112). This, in turn, acts as a driving force for carcinogenesis, highlighting the contribution of IR in initiating and promoting neoplastic transformation.
(ii) Radiation-induced modifications of histones.
IR can cause multiple types of histone modifications. A common IR-induced histone modification is phosphorylation of histone H2AX at serine 139 (γ-H2AX) (113), which is involved in diverse DNA-damage response pathways in mammalian cells through the activation of ATM and ATR (114–117). γH2AX is regarded as a measure of DSBs in DNA and as an early response to IR (118). Capetillo et al. reported that phosphorylated H2BS14 colocalizes with γH2AX loci upon DNA damage by IR (119). In addition, IR—especially UVB—has been reported to induce phosphorylation of H3S10 and H3S28 by the action of kinases, including ERK, p38, and the Src family member Fyn (120, 121). Phosphorylation of these histone moieties is frequently related with the expression of proto-oncogenes, strengthening the conclusion that radiation initiates neoplastic transformation (122).
Apart from phosphorylation, histone methylation also plays a critical role in DNA damage repair and transcriptional modulation. Histone tails can be mono-, di-, or trimethylated at specific lysine and arginine residues by histone methyltransferases, whereas demethylation is carried out by histone demethylases. For instance, laser microirradiation has been reported to load H3K9-specific methyltransferase KMT1A at DBS sites (123). Methylation at K27 of histone H3 (H3K27) is one of the most common modifications at sites of DNA damage induced by UV-laser microirradiation. Di- and trimethylation of H3K27 (H3K27me2 and H3K27me3) is carried out by the Polycomb repressive complex PRC2 via EZH1 and EZH2 (124) (Fig. 1, lower left panel). Targeting EZH2 using shRNAs or the chemical inhibitor GSK126 significantly enhances genotoxicity and promotes radiosensitivity in cancer cells (125). IR also modulates the expression of histone demethylases and histone methyltransferases, thereby impacting the efficacy of cellular responses to DNA damage-induced radiation (126).
(iii) Radiation-induced modulation of noncoding RNA expression.
Several studies have reported alternations in noncoding RNA expression profiles in response to IR (115–118) (Fig. 1, lower left panel). Both lncRNAs and miRNAs play crucial roles in cellular adaptation to radiation stress. In a recent study, Wang et al. identified 14 lncRNAs whose expression is altered in response to gamma irradiation in cervical, breast, and lung cancers (127). The participation of different lncRNAs and miRNAs in mediating radioresistance in cancer cells has been extensively studied by various groups. For instance, TUG1 lncRNA has been reported to induce radioresistance in different types of cancers by modulating miRNA 139-5p (miR-139-5p) and HMGB1 (128). TUG1 knockdown is associated with increased radiosensitivity, suggesting that it can be utilized as a potential therapeutic strategy against different types of solid tumors (129). A recent study in breast cancer reported that exposure to radiation leads to upregulation in expression of lncRNA HOTAIR (130), which in turn stimulated radioresistance by modulating the expression of miR-449b-5p and miR-218 (130, 131). In response to radiation-induced DNA damage, cancer cells rapidly employ DNA repair machinery to avoid cell cycle arrest, which in turn leads to radioresistance. lncRNA LINP1 is overexpressed in multiple cancers and confers resistance to IR and chemotherapeutic drugs by forming polyvalent interactions with Ku, by stabilizing NHEJ complex assembly, and by promoting liquid-phase condensates of break repair proteins thought to be repair foci (132). lncRNA MALAT1 acts in different cancers by inducing DNA repair and inhibiting cell cycle arrest (133, 134). A similar study by Zhang and coworkers reported that lncRNA BOKAS promotes radioresistance by upregulating Wnt1-inducible signaling pathway protein 1 (WISP1) and DNA damage repair (135). lncRNA GAS5 was also reported to suppress radiosensitivity through miR-106b and miR-205-5p (136). The radioresistance of cervical cancer and breast cancer was reported to be mediated by lncRNA NEAT1 and lncRNA LINC00963, respectively (137, 138). NEAT1 enhances cyclin D1 expression through the activity of miR-193b-3p. Apart from these lncRNAs, a number of microRNAs have been reported to be an integrated part of the response to radiation in different cancer models (132). A meta-analysis has reported at least 28 miRNAs that become differentially expressed in response to radiation exposure. Among these, seven miRNAs—miR-150, miR-29a, miR-29b, miR-30c, miR-200b, miR-320a, and miR-30a—may serve as biomarkers due to their elevated levels in serum after exposure to radiation (139). The most widely studied miRNAs in response to radiation are miR-21 and let-7. Studies have shown that let-7 affects RAS as its downstream molecule (140), whereas the downstream target of mir-21 is PTEN (141), indicating the modulation of crucial cellular signaling cascades by these microRNAs in response to radiation stress.
In fact, such RNA-protein interactions directly link chromatin and RNA responses to the outcome to alkylation and IR-induced stress. Conserved from yeast to humans and essential for heterochromatin formation, the vigilin RNA-binding protein with 14 tandemly arranged nonidentical hnRNP K-type homology (KH) domains modulates cell sensitivity to cisplatin- or IR-induced cell death and genomic instability due to defective DNA repair (142). Vigilin connects RNA and DNA responses, as it interacts with the DDR proteins RAD51 and BRCA1, and promotes their recruitment to DSB sites (142). These reports document the response of cancer cells to radiation stress through a multipronged approach that involves different epigenetic regulatory mechanisms.
Adaptation to nutrient deprivation.
Each and every cell modulates their metabolic processes depending upon the extracellular environment and nutrient availability. The intermediates of cellular metabolic processes act as cofactor or substrate of most of the enzymes associated with chromatin modification. Thus, cellular metabolism acts as a link between extracellular environment and chromatin dynamics, regulating genomic accessibility of the transcriptional machinery (143). Fluctuations between the epigenome and metabolism alter the normal physiological balance that ultimately causes complex diseases, such as type II diabetes and cancer.
(i) Epigenetic modifications in response to nutrient deprivation.
In the late 1950s, the discovery of glycolytic enzymes in the nucleus opened a new era of metabolism research. Since then, several studies have documented a fuel-sensing machinery within the cell that regulates the expression of several metabolic enzymes to maintain the metabolome at optimum levels. Nuclear localization of various metabolic enzymes during nutrient stress regulates the activity of different epigenetic modifiers, which ultimately lead to alteration of target gene expression (Fig. 1, lower right panel). Nuclear translocation of these metabolic enzymes depends upon extracellular stimuli and cellular nutrient availability. For instance, upon altered lactate metabolism, phosphorylation of tyrosine 238 of lactate dehydrogenase (LDH) promotes its nuclear import. Within the nucleus, LDH regulates epigenetic modifications by modulating the NAD+ level and activating the SIRT1 deacetylase system (144, 145). Similarly, under low-glucose conditions, hexokinase II (HKII), one of the principal enzymes in glycolysis, becomes phosphorylated by Akt, which promotes translocation to the nuclear compartment (146, 147). A related study in yeast cells showed that a homolog of HKII, HXKII, binds to the transcription factor Mig1 and suppresses the transcription of carbohydrate metabolic enzymes by forming a complex with cyc8 and Tup1 (148). Also, under low-glucose availability, HXKII autophosphorylates and activates Snf1, which relieves repression at the GAL4 promoter by the Mig1 complex and promotes the activity of other transcription factors, such as Adr1 (148). A low glucose level also leads to the nuclear localization of fructose-1,6-bisphosphatase 1 (FBP1), which binds to HIF-1 and HIF-2, thus suppressing transcription of glycolytic enzymes and cell proliferation-related genes (149). Nuclear import of malate dehydrogenase 1 (MDH1) during glucose deprivation promotes apoptosis and p53-mediated cell cycle arrest (150). At normal glucose levels, pyruvate kinase M2 (PKM2) interacts with HIF-1α, inducing the activation of target glycolytic enzyme genes by promoting HIF-1 and p300 binding to their promoters (151). This signaling cascade remains inhibited during glucose starvation. In yeast, Pyk1 (a homolog of PKM2), which is a part of serine-responsive SAM-containing metabolic enzyme complex (SESAME complex), regulates histone H3 phosphorylation depending upon glucose, SAM and serine amino acid levels (152).
(ii) Epigenetic regulation of metabolic enzymes under nutrient stress.
During nutrient deprivation, different metabolic enzymes translocate into the nucleus, where they regulate the expression of different target genes by modulating various epigenetic modifiers. On one hand, acetyl coenzyme A (acetyl-CoA)-producing enzymes such as the pyruvate dehydrogenase complex (PDC), acyl-coenzyme A synthetase short-chain family member 2 (ACSS2), and ATP citrate lyase (ACLY), translocate into the nucleus, increasing the acetyl-CoA pool, which is used by histone acetyltransferases (HATs) for the activation of different target genes (153–155) (Fig. 1, lower right panel). On the other hand, nuclear PDC can form a complex with pyruvate kinase isomerase M2 (PKM2) and p300. This complex then produces acetyl-CoA from phosphoenolpyruvate (PEP), resulting in local increase of acetyl-CoA and histone acetylation by p300 (156). S-Adenosylmethionine (SAM) acts as a methyl group donor that is utilized by different methyltransferases to methylate DNA and histones (157). It is also reported that an increase in SAM pool inside the nucleus inactivates specific target genes (158).
(iii) Epigenetic adaptation to changing nutrient levels.
In cancer cells, epigenetic alterations aimed at reprogramming their metabolic pathways play a crucial role in the response to altered nutrient availability in the tumor milieu. Small molecule metabolites, such as succinate, fumarate, d,l-2-hydroxyglutarate, and β-hydroxybutyrate, inhibit different metabolic enzymes and contribute to tumorigenesis and are therefore considered “oncometabolites” (159). Depending upon the metabolic status of the cell, β-hydroxybutyrate can modulate target gene expression via hyperacetylation of H3K9 and H3K14 (160) and the inhibition of HDACs, thereby regulating cellular growth and survival (161–163).
Activation of glycolysis is part of the program used by cancer cells to support their uncontrolled proliferation. For example, several groups reported a glycolytic flux in hepatocellular carcinoma and glioblastoma by hyperacetylation of the HKII promoter (164–166). Overexpression of lysine demethylase 3A (KDM3A) in bladder cancer promotes glycolysis by activating SLC2A1 (increased GLUT1 expression), phosphoglycerate kinase 1 (PGK1), HKII, lactate dehydrogenase A (LDHA), via demethylation of histone H3K9me2 (167). It was also reported that overexpression of glucose transporter GLUT1 by hypermethylation-mediated silencing of derlin-3 results in elevated glucose uptake to sustain the energy requirement of proliferating cancer cells (168). Alternatively, suppression of gluconeogenesis by hypermethylation of fructose 1,6-biphosphatase (FBP1) was described in liver and colon cancer (169). The epigenetic regulator TCF19, a histone H3K4me3-interacting protein (170), suppresses gluconeogenesis through its selective association with the NuRD complex under high-glucose conditions (171). Interestingly, in hepatocellular carcinoma, TCF19 associates with p53 to form distinct transcriptional complexes that regulate mitochondrial energy metabolism and promote cellular stress adaptation in glucose-mediated stress scenarios (172). It has also been reported that different sirtuins play extensive roles in metabolic reprogramming in cancer cells (Fig. 2). On one hand, SIRT6 causes glycolytic gene activation by augmenting H3K9 acetylation (173, 174) and, on the other hand, SIRT1 suppresses glycolysis by deacetylating HIF-1α and phosphoglycerate mutase 1 (PGAM1), thereby acting as a tumor suppressor (175). A role for SIRT1 as tumor suppressor was also supported by our analysis of TCGA, which showed that SIRT1 mRNA levels were lower in the tumor mass than in matched controls in 13 of 15 tumor types (Fig. 2A). In addition, in 3 of the 33 tumor types comprising TCGA, patients expressing SIRT1 at the lowest levels incurred greater risk of a poor outcome than those with higher mRNA levels of SIRT1 (Fig. 2B). The enzyme pyruvate kinase M2 phosphorylates histone H3 using PEP as the phosphate donor and promotes its acetylation upon stimulation by EGF (176). These interactions increase the expression of cyclin D1 and c-Myc, promoting cell proliferation and cancer progression. All of these reports highlight the altered epigenetic cascades employed by cancer cells to adapt to the steeply reduced nutrient availability in the tumor milieu to achieve cell survival.
Endoplasmic reticulum stress and cancer.
The synthesis of about one-third of all proteins of a eukaryotic cell takes place at the endoplasmic reticulum (ER). In addition to modifying the proteins to target their destination, ER regulates folding and protein quality control. The process of protein folding is highly sensitive to the concentration of calcium ions and the metabolic status of a cell. Protein folding is also influenced by various stimuli, including hypoxia and oxidative stress (177, 178). A disruption in the internal molecular balance of a cell leads to a build-up of incorrectly folded proteins within the ER lumen, altering ER homeostasis and causing ER stress. The elevated burden of misfolded proteins disrupts the balanced interplay between different transcription factors and chromatin dynamics, thereby challenging cellular homeostasis and increasing susceptibility to diabetes, neurodegenerative disorders, heart diseases, inflammatory disorders, and cancer. Misfolded proteins can furthermore impact ROS levels and cell death, as seen for superoxide dismutase mutations that increase folded stability, promote aggregation, and cause the fatal disease amyotrophic lateral sclerosis (179).
(i) Cascade of unfolded protein response.
Upon entering the ER lumen, polypeptide chains are folded into a proper structure by chaperons and modified by different oxidoreductases and glycosylating enzymes (180, 181). Approximately one-third of the polypeptides in the ER lumen fail ER quality control and are removed by the ER-associated degradation (ERAD) machinery (182). During ER stress, the excessive load of misfolded proteins overwhelms the ERAD capability to neutralize the burden and activate the unfolded protein response (UPR), an orchestra of different signaling pathways that rebalance homeostasis (183, 184) (Fig. 1, lower middle panel). Chaperone-binding immunoglobulin protein (BiP) is dissociated from the luminal side of three ER transmembrane proteins: protein kinase RNA-like ER kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1α (IRE1α). This leads to the activation of UPR (185–187). BiP dissociation causes dimerization and autophosphorylation of IRE1α and PERK (188) and coat protein II (COPII)-mediated transport of ATF6 to the Golgi apparatus (189, 190), leading either to cell survival or cell death. If ER stress is excessive, activated IRE1α degrades mRNAs through RNase-like activities, reducing the unfolded protein load by IRE1-dependent decay (RIDD) pathway (191–193), whereas at low ER stress IRE1α triggers the production of spliced X-box binding protein 1 (XBP1s) (194). Moreover, activation of JUN N-terminal kinase (JNK) by IRE1α causes apoptosis by the activation of BAX and BAK (195). XBP1s, in turn, enhances transcription of genes related to protein folding, quality control, ERAD, and phospholipid synthesis (196, 197). In contrast, activated PERK phosphorylates eukaryotic translation initiation factor 2α (eIF2α) and suppresses protein synthesis (198). PERK also induces translation of activating transcription factor 4 (ATF4), which supports transcription of the antioxidant response, autophagy, and apoptosis-related genes (199). ATF4 also stimulates the expression of C/EBP-homologous protein (CHOP), which in turn activates growth arrest and DNA damage-inducible protein 34 (GADD34) and BCL-2 homology 3 (BH3)-only proteins and suppresses BCL-2 (200).
Mahameed et al. indicated the possibility of inducing ER stress as a treatment stratagem against cancer (201). This study has demonstrated the efficacy of a combinatorial approach, including promoting selective retention in the ER (sER) and inhibiting integrated stress response (ISR) as an anticancer regimen. The dual treatment of Nelfinavir (inducer of sER) and ISRIB (inhibitor of ISR) efficiently attenuated the growth of several cell lines, including HepG2, Mel526, HCC827, Mel624, etc. In essence, combinatorial treatment of stress induction, along with chemotherapy, hinders the acquisition of chemoresistance and increases the cellular uptake of chemotherapeutic drugs, leading to an overall reduction of tumors.
(ii) Epigenetic alternations following ER stress.
UPR modulates a magnitude of transcription factors and epigenetic modifiers, ultimately regulating the expression of various target genes. For instance, NF-Y, a transcriptional activator, binds to the ERSE box of ER stress-related gene promoters and recruits ATF6, XBP1s, and other coactivators upon specific epigenetic activation marks, such as H3K4me3 (196, 202). Under normal conditions, heterochromatin protein-like 2 (HPL2) promotes transcriptional suppression of XBP1 by facilitating heterochromatin formation, whereas ER stress inactivates HPL2, thereby activating XBP1 (203). YY1 was reported to interact with ATF6, arginine methyl transferase 1 (PRMT1), and p300 under ER stress conditions (204). Under normal conditions, HDAC1 represses the transcription of UPR genes, while the SAGA-associated factor 29 (SGF29) binds to the histone H3K4me3 mark to maintain these genes in an inactive state. Under ER stress, SGF29 promotes histone H3K14 acetylation at UPR gene promoters (205). NF-Y successively recruits ATF6, YY1, and TBP, which in turn leads to the recruitment of PRMT1 and p300. PRMT1 and p300 methylate and acetylate histone H4, respectively, causing transcriptional activation of UPR-responsive genes (204).
(iii) Epigenetic regulation of ER stress and its connection to cancer.
Mutation, nutrient deprivation, and ATP deficiency within cancer cells increase the number of incorrectly folded proteins within the ER lumen, leading to ER stress. During glucose deficiency, the PERK pathway activates several proangiogenic factors, such as VEGF, FGF2, and IL-6, and suppresses expression of angiogenic inhibitor proteins, including THBS1, CXCL14, and CXCL10 (206). Activated PERK also promotes miR-211 production, which in turn represses CHOP expression by binding to its nascent mRNAs during transcription and increases histone H3K27me3 mark (207). The transcription factor CHOP is a member of the C/EBP family and is reported as a tumor suppressor (208). CHOP is reported to be activated by UPR and induces apoptosis in cancer cells (209) and has also been reported to promote brain metastasis via miR-708 expression (192).
ER stress responsive factors are correlated with tumor progression (193–195). For example, it has been reported that, during hypoxia, HIF-1α forms a complex with XBP1s to prevent apoptosis (210). IRE1α degrades transcription repressor protein period1, thereby resulting in the expression of chemokine (C-X-C motif) ligand 3 (CXCL3), leading to tumor development (211). Ubiquitination of H2A prevents RNA polymerase II from transcribing the proapoptotic gene NOXA in cancer cells. However, the upregulation of ATF3 and ATF4 activates the expression of NOXA by disrupting H2A ubiquitination (212). These and other studies collectively point toward the association of ER stress with cancer development, survival, and metastasis.
UTILIZING STRESS IN THE THERAPEUTIC REGIMEN OF ANTICANCER THERAPY
As evidenced in discussion above, the maintenance of homeostasis and its adaptive responses to stress will largely determine whether the cancer cells can survive a stressful stimulus or will undergo apoptosis. With this in mind, the harnessing and combination of stress may provide powerful therapeutic opportunities. Most current anticancer therapeutic regimens utilize the induction of stress as an approach to target cancer cells. Chemotherapeutic regimens such as alkylating agents and intercalating agents induce genotoxic stress to target tumor cells. Indeed, the first implemented anticancer drugs were alkylating agents, which are still among the most frequently used chemotherapeutic drugs; however, we are still learning their mechanisms of action and impact on RNA, which may affect how they can be best combined with other types of stress (213). Intriguingly, some of the RNA modifications produced by alkylating agents are also elicited by physiological processes. Thus, alkylation altering epigenetic marks merits more investigation (214).
As a routine first-line treatment against most aggressive carcinomas, chemotherapeutic drugs are often combined with radiotherapy to maximize genotoxic damage. For instance, cotreatment with temozolomide and radiotherapy is a standardized procedure against glioblastoma (215), increasing the median survival of patients (216). Chemotherapeutic drugs and radiation can also increase ROS levels, inducing oxidative stress and subsequent DNA damage. Cancer cells inherently display heightened ROS levels and increased tolerance to ROS (217). When levels of ROS are further elevated in cancer cells, they disrupt redox homeostasis, overriding the defensive antioxidant system of cancer cells and resulting in cell death. The induction of programmed cancer cell death by strategically elevating the levels of ROS is the principal mechanism behind many anticancer agents. Radiation therapy and most chemotherapeutic agents lead to the induction of intracellular ROS levels and stimulation of apoptosis via both extrinsic and intrinsic pathways (218).
Targeted therapies such as PDGFR inhibitor, EGFR inhibitor, BRAF inhibitor, and several monoclonal antibodies stimulate apoptosis by inducing oxidative stress (216, 219–221). In Hodgkin’s lymphoma and brain cancers, procarbazine was reported to induce irreparable oxidative DNA damage (222). In breast cancer, bladder cancer, colon cancer, and acute lymphocytic leukemia, the routinely used chemotherapeutic agents doxorubicin and 5-fluorouracil have been reported to produce ROS via p53-dependent or -independent pathways (223, 224).
In general, we are increasingly finding that epigenetics and metabolomics are interwoven with DNA replication and repair in ways that determine adaptions to stress and cancer cell fate. Hence, important new targeted therapies are coming from molecular targets in the DDR. Building upon the success of inhibitors to the PARP1 poly(ADP-ribose) (PAR) polymerase, inhibitors to the PAR glyocohydrolase (PARG) needed to hydrolyze the “cloud” of PAR into mononucleotide ADP-ribose (ADPr) are being tested (225, 226). Furthermore, excessive or unhydrolyzed PAR depletes the key metabolite NAD+ and triggers cell death by parthanatos by the apoptosis-inducing factor (AIF) via an allosteric mechanism that provides an interesting molecular cancer target. AIF enables mitochondrial regulation during sustained changes in NAD+ levels in response to diet, aging, or chronic, disease-induced PARP-1 hyperactivation, complementing NAD+-driven transcriptional regulation of mitochondrial biogenesis (227). Separately and in combination with alkylating and cross-linking agents that target replicative stress, inhibitors to replication stress responses such as the replication stress kinase ATR are being examined in clinical trials, making the EXO5 nuclease responsible for ATR-mediated replication restart an attractive cancer target (228, 229).
Epigenetic changes can result in what appears to be the biggest block to replication (and thus to cancer cell proliferation), which is a collision with transcription. Indeed, the nucleotide excision repair (NER) complex, which provides resistance to alkylation therapy by repairing bulky DNA lesions, may have evolved first for transcription-coupled NER from evolutionary pressure to resolve arrested transcription blocks to DNA replication (230). These considerations suggest that epigenetic transcriptional changes may be key to replication collisions that could be manipulated to promote instability or cell death in oncogenesis.
Unfortunately, acquisition of resistance toward chemotherapy, radiation therapy, and targeted therapies such as PARP inhibitors pose a major challenge to the management of advanced cancers. This has led to development of complementary strategies to target cancer cells in a multipronged therapeutic approach; one example includes oncothermia, a method of regulated hyperthermia where the temperature in the extracellular tumor liquid is increased locally by applying deep heat. A recent study in colorectal cancer reported the upregulation of the proapoptotic markers Bax and Puma and the downregulation of antiapoptotic Xiap, Bcl-2, and Bcl-xl markers by oncothermia treatment (231). Another study in the hepatocellular carcinoma cell line HepG2 indicated that oncothermia leads to apoptosis by increasing the levels of caspase-3, −8, and −9 (232). In summary, the combinatorial treatment of oncothermic stress induction, along with chemotherapy, appears to hinder the acquisition of chemoresistance and to increase the cellular uptake of chemotherapeutic drugs, leading to an overall reduction of tumor mass (204).
Since the induction of stress can serve as a potential approach to efficaciously target cancer cells, the overall understanding of the epigenetic landscape involved in stress response can unveil a plethora of new possibilities in devising anticancer regimens. In this context, CRISPR/Cas9 technology may prove to be of unparallel potential to engineer epigenome in a way so that the cancer cells can be efficiently manipulated to succumb to stress-mediated apoptosis, ultimately leading to tumor regression (233, 234). Combinatorial strategies, including the induction of stress and the modulation of epigenome to attenuate stress resistance, can act as a dual strategy which can prove to be a potential highlight in the future avenue of anticancer research.
Overall, stress offers opportunities for amplification of cancer cell killing by unbalancing DDR. This stress-DDR rebalancing can result both in increased damage and in combined transcriptional and replicational stress that can particularly target cancer cells, as they have intrinsic elements of transcription and replication stress. This is seen, for example, with ROS-causing radiation therapy, hyperthermia, metabolite NAD+ decreases during excess PARylation, and epigenetic RNA alterations in alkylation therapy. However, this approach will require a mechanistic understanding of the interplay between stress and DDR. For example, DNA damage in germ cells evokes elevated resistance to heat and oxidative stress by induction of inflammation and innate immune response. This can lead to activation of the ubiquitin-proteasome system (UPS) in somatic tissues, which confers enhanced proteostasis and systemic stress resistance (235). Yet, with the mechanistic knowledge of stress response networks, we can begin to better predict therapeutic effects and the prognosis of cancer patients by monitoring the related stress pathways, aiming for more effective and individualized treatment to improve the therapeutic efficacy of current therapies and their combination with targeted stress, such as oncothermia. Going forward, we expect to see more strategic applications harnessing the stress responses outlined here in order to overcome resistance toward chemotherapy, radiation therapy, and targeted molecular therapies.
ACKNOWLEDGMENTS
We thank the members of our laboratories for insightful discussions. Regretfully, many excellent papers could not be included here due to space limitations and our focus on results with which we are most familiar for the exemplary work cited within this broad area.
This research was supported by the National Institutes of Health (NIH; grants P01 CA092584, R35 CA220430, and P30 GM124169) and the Robert A. Welch Chemistry Chair and Cancer Prevention and Research Institute of Texas (CPRIT; grant RP180813) to J.A.T. This study used computing resources from the Texas Advanced Computing Center (TACC) at the University of Texas in Austin, TX. C.D. acknowledges support from Basic and Applied Research in Biophysics and Material Science (RSI 4002) by the Department of Atomic Energy (DAE), Government of India, SwarnaJayanti Fellowship (DST/SJF/LSA-02/2017-18), Department of Science and Technology, and an S. Ramachandran National Bioscience Award for Career Development 2019 (BT/HRD-NBA-NWB/38/2019-20), Department of Biotechnology.
Contributor Information
Tej K. Pandita, Email: Tej.Pandita@BCM.edu.
Chandrima Das, Email: chandrima.das@saha.ac.in.
REFERENCES
- 1.Garcia-Martinez L, Zhang Y, Nakata Y, Chan HL, Morey L. 2021. Epigenetic mechanisms in breast cancer therapy and resistance. Nat Commun 12:1786. 10.1038/s41467-021-22024-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Di Nicolantonio F, Mercer SJ, Knight LA, Gabriel FG, Whitehouse PA, Sharma S, Fernando A, Glaysher S, Di Palma S, Johnson P, Somers SS, Toh S, Higgins B, Lamont A, Gulliford T, Hurren J, Yiangou C, Cree IA. 2005. Cancer cell adaptation to chemotherapy. BMC Cancer 5:78. 10.1186/1471-2407-5-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhao RZ, Jiang S, Zhang L, Yu ZB. 2019. Mitochondrial electron transport chain, ROS generation and uncoupling (review). Int J Mol Med 44:3–15. 10.3892/ijmm.2019.4188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Quinlan CL, Orr AL, Perevoshchikova IV, Treberg JR, Ackrell BA, Brand MD. 2012. Mitochondrial complex II can generate reactive oxygen species at high rates in both the forward and reverse reactions. J Biol Chem 287:27255–27264. 10.1074/jbc.M112.374629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sies H. 1997. Oxidative stress: oxidants and antioxidants. Exp Physiol 82:291–295. 10.1113/expphysiol.1997.sp004024. [DOI] [PubMed] [Google Scholar]
- 6.Zhang J, Zheng YG. 2016. SAM/SAH analogs as versatile tools for SAM-dependent methyltransferases. ACS Chem Biol 11:583–597. 10.1021/acschembio.5b00812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bansal A, Simon MC. 2018. Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol 217:2291–2298. 10.1083/jcb.201804161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pajares MA, Duran C, Corrales F, Pliego MM, Mato JM. 1992. Modulation of rat liver S-adenosylmethionine synthetase activity by glutathione. J Biol Chem 267:17598–17605. 10.1016/S0021-9258(19)37084-X. [DOI] [PubMed] [Google Scholar]
- 9.Colin DJ, Limagne E, Ragot K, Lizard G, Ghiringhelli F, Solary E, Chauffert B, Latruffe N, Delmas D. 2014. The role of reactive oxygen species and subsequent DNA-damage response in the emergence of resistance towards resveratrol in colon cancer models. Cell Death Dis 5:e1533. 10.1038/cddis.2014.486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Achanta G, Huang P. 2004. Role of p53 in sensing oxidative DNA damage in response to reactive oxygen species-generating agents. Cancer Res 64:6233–6239. 10.1158/0008-5472.CAN-04-0494. [DOI] [PubMed] [Google Scholar]
- 11.Cooke MS, Evans MD, Dizdaroglu M, Lunec J. 2003. Oxidative DNA damage: mechanisms, mutation, and disease. FASEB J 17:1195–1214. 10.1096/fj.02-0752rev. [DOI] [PubMed] [Google Scholar]
- 12.O’Hagan HM, Wang W, Sen S, Destefano Shields C, Lee SS, Zhang YW, Clements EG, Cai Y, Van Neste L, Easwaran H, Casero RA, Sears CL, Baylin SB. 2011. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell 20:606–619. 10.1016/j.ccr.2011.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Waris G, Ahsan H. 2006. Reactive oxygen species: role in the development of cancer and various chronic conditions. J Carcinog 5:14. 10.1186/1477-3163-5-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mahalingaiah PK, Ponnusamy L, Singh KP. 2017. Oxidative stress-induced epigenetic changes associated with malignant transformation of human kidney epithelial cells. Oncotarget 8:11127–11143. 10.18632/oncotarget.12091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Garcia-Guede A, Vera O, Ibanez-de-Caceres I. 2020. When oxidative stress meets epigenetics: implications in cancer development. Antioxidants (Basel) 9:468. 10.3390/antiox9060468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nishida N, Arizumi T, Takita M, Kitai S, Yada N, Hagiwara S, Inoue T, Minami Y, Ueshima K, Sakurai T, Kudo M. 2013. Reactive oxygen species induce epigenetic instability through the formation of 8-hydroxydeoxyguanosine in human hepatocarcinogenesis. Dig Dis 31:459–466. 10.1159/000355245. [DOI] [PubMed] [Google Scholar]
- 17.Musarrat J, Arezina-Wilson J, Wani AA. 1996. Prognostic and aetiological relevance of 8-hydroxyguanosine in human breast carcinogenesis. Eur J Cancer 32:1209–1214. 10.1016/0959-8049(96)00031-7. [DOI] [PubMed] [Google Scholar]
- 18.Barciszewska A-M, Giel-Pietraszuk M, Perrigue PM, Naskręt-Barciszewska M. 2019. Total DNA methylation changes reflect random oxidative DNA damage in gliomas. Cells 8:1065. 10.3390/cells8091065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Perez S, Rius-Perez S, Tormos AM, Finamor I, Nebreda AR, Talens-Visconti R, Sastre J. 2018. Age-dependent regulation of antioxidant genes by p38α MAPK in the liver. Redox Biol 16:276–284. 10.1016/j.redox.2018.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. 2010. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49:1603–1616. 10.1016/j.freeradbiomed.2010.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Inoue I, Goto S, Matsunaga T, Nakajima T, Awata T, Hokari S, Komoda T, Katayama S. 2001. The ligands/activators for peroxisome proliferator-activated receptor alpha (PPARα) and PPARγ increase Cu2+,Zn2+-superoxide dismutase and decrease p22phox message expressions in primary endothelial cells. Metabolism 50:3–11. 10.1053/meta.2001.19415. [DOI] [PubMed] [Google Scholar]
- 22.Jung JE, Kim GS, Narasimhan P, Song YS, Chan PH. 2009. Regulation of Mn-superoxide dismutase activity and neuroprotection by STAT3 in mice after cerebral ischemia. J Neurosci 29:7003–7014. 10.1523/JNEUROSCI.1110-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lim SO, Gu JM, Kim MS, Kim HS, Park YN, Park CK, Cho JW, Park YM, Jung G. 2008. Epigenetic changes induced by reactive oxygen species in hepatocellular carcinoma: methylation of the E-cadherin promoter. Gastroenterology 135:2128–2140. 10.1053/j.gastro.2008.07.027. [DOI] [PubMed] [Google Scholar]
- 24.Kang KA, Zhang R, Kim GY, Bae SC, Hyun JW. 2012. Epigenetic changes induced by oxidative stress in colorectal cancer cells: methylation of tumor suppressor RUNX3. Tumour Biol 33:403–412. 10.1007/s13277-012-0322-6. [DOI] [PubMed] [Google Scholar]
- 25.Sharma GG, So S, Gupta A, Kumar R, Cayrou C, Avvakumov N, Bhadra U, Pandita RK, Porteus MH, Chen DJ, Cote J, Pandita TK. 2010. MOF and histone H4 acetylation at lysine 16 are critical for DNA damage response and double-strand break repair. Mol Cell Biol 30:3582–3595. 10.1128/MCB.01476-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Horikoshi N, Sharma D, Leonard F, Pandita RK, Charaka VK, Hambarde S, Horikoshi NT, Gaur Khaitan P, Chakraborty S, Cote J, Godin B, Hunt CR, Pandita TK. 2019. Preexisting H4K16ac levels in euchromatin drive DNA repair by homologous recombination in S-phase. Commun Biol 2:253. 10.1038/s42003-019-0498-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wongpaiboonwattana W, Tosukhowong P, Dissayabutra T, Mutirangura A, Boonla C. 2013. Oxidative stress induces hypomethylation of LINE-1 and hypermethylation of the RUNX3 promoter in a bladder cancer cell line. Asian Pac J Cancer Prev 14:3773–3778. 10.7314/apjcp.2013.14.6.3773. [DOI] [PubMed] [Google Scholar]
- 28.Dhani N, Fyles A, Hedley D, Milosevic M. 2015. The clinical significance of hypoxia in human cancers. Semin Nucl Med 45:110–121. 10.1053/j.semnuclmed.2014.11.002. [DOI] [PubMed] [Google Scholar]
- 29.Vaupel P. 2008. Hypoxia and aggressive tumor phenotype: implications for therapy and prognosis. Oncologist 13(Suppl 3):21–26. 10.1634/theoncologist.13-S3-21. [DOI] [PubMed] [Google Scholar]
- 30.Hockel M, Vaupel P. 2001. Biological consequences of tumor hypoxia. Semin Oncol 28:36–41. 10.1016/S0093-7754(01)90211-8. [DOI] [PubMed] [Google Scholar]
- 31.Weidemann A, Johnson RS. 2008. Biology of HIF-1α. Cell Death Differ 15:621–627. 10.1038/cdd.2008.12. [DOI] [PubMed] [Google Scholar]
- 32.Semenza GL, Wang GL. 1992. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 12:5447–5454. 10.1128/MCB.12.12.5447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Jiang BH, Semenza GL, Bauer C, Marti HH. 1996. Hypoxia-inducible factor 1 levels vary exponentially over a physiologically relevant range of O2 tension. Am J Physiol 271:C1172–C1180. 10.1152/ajpcell.1996.271.4.C1172. [DOI] [PubMed] [Google Scholar]
- 34.Qin J, Liu Y, Lu Y, Liu M, Li M, Li J, Wu L. 2017. Hypoxia-inducible factor 1α promotes cancer stem cell-like properties in human ovarian cancer cells by upregulating SIRT1 expression. Sci Rep 7:10592. 10.1038/s41598-017-09244-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mimeault M, Batra SK. 2013. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J Cell Mol Med 17:30–54. 10.1111/jcmm.12004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM, Johnson RS. 2000. Hypoxia-inducible factor-1α is a positive factor in solid tumor growth. Cancer Res 60:4010–4015. [PubMed] [Google Scholar]
- 37.Ryan HE, Lo J, Johnson RS. 1998. HIF-1α is required for solid tumor formation and embryonic vascularization. EMBO J 17:3005–3015. 10.1093/emboj/17.11.3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, Semenza GL. 1996. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 16:4604–4613. 10.1128/MCB.16.9.4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bruick RK, McKnight SL. 2001. A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294:1337–1340. 10.1126/science.1066373. [DOI] [PubMed] [Google Scholar]
- 40.Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, Pavletich N, Chau V, Kaelin WG. 2000. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol 2:423–427. 10.1038/35017054. [DOI] [PubMed] [Google Scholar]
- 41.Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ. 1999. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399:271–275. 10.1038/20459. [DOI] [PubMed] [Google Scholar]
- 42.Huang LE, Gu J, Schau M, Bunn HF. 1998. Regulation of hypoxia-inducible factor 1alpha is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc Natl Acad Sci USA 95:7987–7992. 10.1073/pnas.95.14.7987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cheng J, Kang X, Zhang S, Yeh ET. 2007. SUMO-specific protease 1 is essential for stabilization of HIF-1α during hypoxia. Cell 131:584–595. 10.1016/j.cell.2007.08.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kietzmann T, Mennerich D, Dimova EY. 2016. Hypoxia-inducible factors (HIFs) and phosphorylation: impact on stability, localization, and transactivity. Front Cell Dev Biol 4:11. 10.3389/fcell.2016.00011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Duyndam MC, Hulscher ST, van der Wall E, Pinedo HM, Boven E. 2003. Evidence for a role of p38 kinase in hypoxia-inducible factor 1-independent induction of vascular endothelial growth factor expression by sodium arsenite. J Biol Chem 278:6885–6895. 10.1074/jbc.M206320200. [DOI] [PubMed] [Google Scholar]
- 46.Jeong JW, Bae MK, Ahn MY, Kim SH, Sohn TK, Bae MH, Yoo MA, Song EJ, Lee KJ, Kim KW. 2002. Regulation and destabilization of HIF-1α by ARD1-mediated acetylation. Cell 111:709–720. 10.1016/s0092-8674(02)01085-1. [DOI] [PubMed] [Google Scholar]
- 47.Schmitt AM, Schmid S, Rudolph T, Anlauf M, Prinz C, Kloppel G, Moch H, Heitz PU, Komminoth P, Perren A. 2009. VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer 16:1219–1227. 10.1677/ERC-08-0297. [DOI] [PubMed] [Google Scholar]
- 48.Koslowski M, Luxemburger U, Tureci O, Sahin U. 2011. Tumor-associated CpG demethylation augments hypoxia-induced effects by positive autoregulation of HIF-1α. Oncogene 30:876–882. 10.1038/onc.2010.481. [DOI] [PubMed] [Google Scholar]
- 49.Kim I, Park JW. 2020. Hypoxia-driven epigenetic regulation in cancer progression: a focus on histone methylation and its modifying enzymes. Cancer Lett 489:41–49. 10.1016/j.canlet.2020.05.025. [DOI] [PubMed] [Google Scholar]
- 50.Casciello F, Al-Ejeh F, Kelly G, Brennan DJ, Ngiow SF, Young A, Stoll T, Windloch K, Hill MM, Smyth MJ, Gannon F, Lee JS. 2017. G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis. Proc Natl Acad Sci USA 114:7077–7082. 10.1073/pnas.1618706114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Li Y, Guo X, Sun L, Xiao J, Su S, Du S, Li Z, Wu S, Liu W, Mo K, Xia S, Chang YJ, Denis D, Tao YX. 2020. N(6)-methyladenosine demethylase FTO contributes to neuropathic pain by stabilizing G9a expression in primary sensory neurons. Adv Sci 7:1902402. 10.1002/advs.201902402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lee JS, Kim Y, Kim IS, Kim B, Choi HJ, Lee JM, Shin HJ, Kim JH, Kim JY, Seo SB, Lee H, Binda O, Gozani O, Semenza GL, Kim M, Kim KI, Hwang D, Baek SH. 2010. Negative regulation of hypoxic responses via induced Reptin methylation. Mol Cell 39:71–85. 10.1016/j.molcel.2010.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lee JS, Kim Y, Bhin J, Shin HJ, Nam HJ, Lee SH, Yoon JB, Binda O, Gozani O, Hwang D, Baek SH. 2011. Hypoxia-induced methylation of a pontin chromatin remodeling factor. Proc Natl Acad Sci USA 108:13510–13515. 10.1073/pnas.1106106108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Perez-Perri JI, Dengler VL, Audetat KA, Pandey A, Bonner EA, Urh M, Mendez J, Daniels DL, Wappner P, Galbraith MD, Espinosa JM. 2016. The TIP60 complex is a conserved coactivator of HIF-1α. Cell Rep 16:37–47. 10.1016/j.celrep.2016.05.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Schoepflin ZR, Shapiro IM, Risbud MV. 2016. Class I and IIa HDACs mediate HIF-1α stability through PHD2-dependent mechanism, while HDAC6, a class IIb member, promotes HIF-1α transcriptional activity in nucleus pulposus cells of the intervertebral disc. J Bone Miner Res 31:1287–1299. 10.1002/jbmr.2787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Geng H, Liu Q, Xue C, David LL, Beer TM, Thomas GV, Dai MS, Qian DZ. 2012. HIF-1α protein stability is increased by acetylation at lysine 709. J Biol Chem 287:35496–35505. 10.1074/jbc.M112.400697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lim JH, Lee YM, Chun YS, Chen J, Kim JE, Park JW. 2010. Sirtuin 1 modulates cellular responses to hypoxia by deacetylating hypoxia-inducible factor 1α. Mol Cell 38:864–878. 10.1016/j.molcel.2010.05.023. [DOI] [PubMed] [Google Scholar]
- 58.Schumacker PT. 2011. SIRT3 controls cancer metabolic reprogramming by regulating ROS and HIF. Cancer Cell 19:299–300. 10.1016/j.ccr.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wang Y, Chen W, Lian J, Zhang H, Yu B, Zhang M, Wei F, Wu J, Jiang J, Jia Y, Mo F, Zhang S, Liang X, Mou X, Tang J. 2020. The lncRNA PVT1 regulates nasopharyngeal carcinoma cell proliferation via activating the KAT2A acetyltransferase and stabilizing HIF-1α. Cell Death Differ 27:695–710. 10.1038/s41418-019-0381-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wen X, Liu X, Mao YP, Yang XJ, Wang YQ, Zhang PP, Lei Y, Hong XH, He QM, Ma J, Liu N, Li YQ. 2018. Long noncoding RNA DANCR stabilizes HIF-1α and promotes metastasis by interacting with NF90/NF45 complex in nasopharyngeal carcinoma. Theranostics 8:5676–5689. 10.7150/thno.28538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lee S, Hallis SP, Jung KA, Ryu D, Kwak MK. 2019. Impairment of HIF-1α-mediated metabolic adaption by NRF2-silencing in breast cancer cells. Redox Biol 24:101210. 10.1016/j.redox.2019.101210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zeng Z, Xu FY, Zheng H, Cheng P, Chen QY, Ye Z, Zhong JX, Deng SJ, Liu ML, Huang K, Li Q, Li W, Hu YH, Wang F, Wang CY, Zhao G. 2019. LncRNA-MTA2TR functions as a promoter in pancreatic cancer via driving deacetylation-dependent accumulation of HIF-1α. Theranostics 9:5298–5314. 10.7150/thno.34559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zheng Y, Wu C, Yang J, Zhao Y, Jia H, Xue M, Xu D, Yang F, Fu D, Wang C, Hu B, Zhang Z, Li T, Yan S, Wang X, Nelson PJ, Bruns C, Qin L, Dong Q. 2020. Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer. Signal Transduct Target Ther 5:53. 10.1038/s41392-020-0146-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wu MZ, Tsai YP, Yang MH, Huang CH, Chang SY, Chang CC, Teng SC, Wu KJ. 2011. Interplay between HDAC3 and WDR5 is essential for hypoxia-induced epithelial-mesenchymal transition. Mol Cell 43:811–822. 10.1016/j.molcel.2011.07.012. [DOI] [PubMed] [Google Scholar]
- 65.Zaidi SK, Frietze SE, Gordon JA, Heath JL, Messier T, Hong D, Boyd JR, Kang M, Imbalzano AN, Lian JB, Stein JL, Stein GS. 2017. Bivalent epigenetic control of oncofetal gene expression in cancer. Mol Cell Biol 37. 10.1128/MCB.00352-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Engin K. 1996. Biological rationale and clinical experience with hyperthermia. Control Clin Trials 17:316–342. 10.1016/0197-2456(95)00078-x. [DOI] [PubMed] [Google Scholar]
- 67.Hunt CR, Pandita RK, Laszlo A, Higashikubo R, Agarwal M, Kitamura T, Gupta A, Rief N, Horikoshi N, Baskaran R, Lee JH, Lobrich M, Paull TT, Roti Roti JL, Pandita TK. 2007. Hyperthermia activates a subset of ataxia-telangiectasia mutated effectors independent of DNA strand breaks and heat shock protein 70 status. Cancer Res 67:3010–3017. 10.1158/0008-5472.CAN-06-4328. [DOI] [PubMed] [Google Scholar]
- 68.Ritossa F. 1962. A new puffing pattern induced by temperature shock and DNP in drosophila. Experientia 18:571–573. 10.1007/BF02172188. [DOI] [Google Scholar]
- 69.Hartl FU, Bracher A, Hayer-Hartl M. 2011. Molecular chaperones in protein folding and proteostasis. Nature 475:324–332. 10.1038/nature10317. [DOI] [PubMed] [Google Scholar]
- 70.Joutsen J, Sistonen L. 2019. Tailoring of proteostasis networks with heat shock factors. Cold Spring Harb Perspect Biol 11:a034066. 10.1101/cshperspect.a034066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Abane R, Mezger V. 2010. Roles of heat shock factors in gametogenesis and development. FEBS J 277:4150–4172. 10.1111/j.1742-4658.2010.07830.x. [DOI] [PubMed] [Google Scholar]
- 72.Jin X, Eroglu B, Moskophidis D, Mivechi NF. 2018. Targeted deletion of Hsf1, −2, and −4 genes in mice. Methods Mol Biol 1709:1–22. 10.1007/978-1-4939-7477-1_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dai C, Whitesell L, Rogers AB, Lindquist S. 2007. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell 130:1005–1018. 10.1016/j.cell.2007.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Xi C, Hu Y, Buckhaults P, Moskophidis D, Mivechi NF. 2012. Heat shock factor Hsf1 cooperates with ErbB2 (Her2/Neu) protein to promote mammary tumorigenesis and metastasis. J Biol Chem 287:35646–35657. 10.1074/jbc.M112.377481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Min JN, Huang L, Zimonjic DB, Moskophidis D, Mivechi NF. 2007. Selective suppression of lymphomas by functional loss of Hsf1 in a p53-deficient mouse model for spontaneous tumors. Oncogene 26:5086–5097. 10.1038/sj.onc.1210317. [DOI] [PubMed] [Google Scholar]
- 76.Dong B, Jaeger AM, Thiele DJ. 2019. Inhibiting heat shock factor 1 in cancer: a unique therapeutic opportunity. Trends Pharmacol Sci 40:986–1005. 10.1016/j.tips.2019.10.008. [DOI] [PubMed] [Google Scholar]
- 77.Brusselaers N, Ekwall K, Durand-Dubief M. 2019. Copy number of 8q24.3 drives HSF1 expression and patient outcome in cancer: an individual patient data meta-analysis. Hum Genomics 13:54. 10.1186/s40246-019-0241-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Li J, Song P, Jiang T, Dai D, Wang H, Sun J, Zhu L, Xu W, Feng L, Shin VY, Morrison H, Wang X, Jin H. 2018. Heat shock factor 1 epigenetically stimulates glutaminase-1-dependent mTOR activation to promote colorectal carcinogenesis. Mol Ther 26:1828–1839. 10.1016/j.ymthe.2018.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhong YH, Cheng HZ, Peng H, Tang SC, Wang P. 2016. Heat shock factor 2 is associated with the occurrence of lung cancer by enhancing the expression of heat shock proteins. Oncol Lett 12:5106–5112. 10.3892/ol.2016.5368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Jin X, Eroglu B, Cho W, Yamaguchi Y, Moskophidis D, Mivechi NF. 2012. Inactivation of heat shock factor Hsf4 induces cellular senescence and suppresses tumorigenesis in vivo. Mol Cancer Res 10:523–534. 10.1158/1541-7786.MCR-11-0530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Takahashi A, Mori E, Somakos GI, Ohnishi K, Ohnishi T. 2008. Heat induces γH2AX foci formation in mammalian cells. Mutat Res 656:88–92. 10.1016/j.mrgentox.2008.07.012. [DOI] [PubMed] [Google Scholar]
- 82.Takahashi A, Mori E, Su X, Nakagawa Y, Okamoto N, Uemura H, Kondo N, Noda T, Toki A, Ejima Y, Chen DJ, Ohnishi K, Ohnishi T. 2010. ATM is the predominant kinase involved in the phosphorylation of histone H2AX after heating. J Radiat Res 51:417–422. 10.1269/jrr.10015. [DOI] [PubMed] [Google Scholar]
- 83.Takahashi A, Mori E, Ohnishi T. 2010. The foci of DNA double strand break-recognition proteins localize with γH2AX after heat treatment. J Radiat Res 51:91–95. 10.1269/jrr.09111. [DOI] [PubMed] [Google Scholar]
- 84.Oei AL, Vriend LE, Crezee J, Franken NA, Krawczyk PM. 2015. Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all. Radiat Oncol 10:165. 10.1186/s13014-015-0462-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bergs JW, Krawczyk PM, Borovski T, ten Cate R, Rodermond HM, Stap J, Medema JP, Haveman J, Essers J, van Bree C, Stalpers LJ, Kanaar R, Aten JA, Franken NA. 2013. Inhibition of homologous recombination by hyperthermia shunts early double strand break repair to non-homologous end-joining. DNA Repair (Amst) 12:38–45. 10.1016/j.dnarep.2012.10.008. [DOI] [PubMed] [Google Scholar]
- 86.Beck BD, Dynlacht JR. 2001. Heat-induced aggregation of XRCC5 (Ku80) in nontolerant and thermotolerant cells. Radiat Res 156:767–774. 10.1667/0033-7587(2001)156[0767:HIAOXK]2.0.CO;2. [DOI] [PubMed] [Google Scholar]
- 87.Ihara M, Suwa A, Komatsu K, Shimasaki T, Okaichi K, Hendrickson EA, Okumura Y. 1999. Heat sensitivity of double-stranded DNA-dependent protein kinase (DNA-PK) activity. Int J Radiat Biol 75:253–258. 10.1080/095530099140717. [DOI] [PubMed] [Google Scholar]
- 88.Burgman P, Ouyang H, Peterson S, Chen DJ, Li GC. 1997. Heat inactivation of Ku autoantigen: possible role in hyperthermic radiosensitization. Cancer Res 57:2847–2850. [PubMed] [Google Scholar]
- 89.Matsumoto Y, Suzuki N, Sakai K, Morimatsu A, Hirano K, Murofushi H. 1997. A possible mechanism for hyperthermic radiosensitization mediated through hyperthermic lability of Ku subunits in DNA-dependent protein kinase. Biochem Biophys Res Commun 234:568–572. 10.1006/bbrc.1997.6689. [DOI] [PubMed] [Google Scholar]
- 90.Ihara M, Takeshita S, Okaichi K, Okumura Y, Ohnishi T. 2014. Heat exposure enhances radiosensitivity by depressing DNA-PK kinase activity during double strand break repair. Int J Hyperthermia 30:102–109. 10.3109/02656736.2014.887793. [DOI] [PubMed] [Google Scholar]
- 91.Hammel M, Tainer JA. 2021. X-ray scattering reveals disordered linkers and dynamic interfaces in complexes and mechanisms for DNA double-strand break repair impacting cell and cancer biology. Protein Sci 30:1735–1756. 10.1002/pro.4133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Krawczyk PM, Eppink B, Essers J, Stap J, Rodermond H, Odijk H, Zelensky A, van Bree C, Stalpers LJ, Buist MR, Soullie T, Rens J, Verhagen HJ, O’Connor MJ, Franken NA, Ten Hagen TL, Kanaar R, Aten JA. 2011. Mild hyperthermia inhibits homologous recombination, induces BRCA2 degradation, and sensitizes cancer cells to poly (ADP-ribose) polymerase-1 inhibition. Proc Natl Acad Sci USA 108:9851–9856. 10.1073/pnas.1101053108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Gerashchenko BI, Gooding G, Dynlacht JR. 2010. Hyperthermia alters the interaction of proteins of the Mre11 complex in irradiated cells. Cytometry A 77:940–952. 10.1002/cyto.a.20955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Xu M, Myerson RJ, Xia Y, Whitehead T, Moros EG, Straube WL, Roti JL. 2007. The effects of 41°C hyperthermia on the DNA repair protein, MRE11, correlate with radiosensitization in four human tumor cell lines. Int J Hyperthermia 23:343–351. 10.1080/02656730701383007. [DOI] [PubMed] [Google Scholar]
- 95.Seno JD, Dynlacht JR. 2004. Intracellular redistribution and modification of proteins of the Mre11/Rad50/Nbs1 DNA repair complex following irradiation and heat shock. J Cell Physiol 199:157–170. 10.1002/jcp.10475. [DOI] [PubMed] [Google Scholar]
- 96.Zhu WG, Seno JD, Beck BD, Dynlacht JR. 2001. Translocation of MRE11 from the nucleus to the cytoplasm as a mechanism of radiosensitization by heat. Radiat Res 156:95–102. 10.1667/0033-7587(2001)156[0095:TOMFTN]2.0.CO;2. [DOI] [PubMed] [Google Scholar]
- 97.Gupta A, Sharma GG, Young CS, Agarwal M, Smith ER, Paull TT, Lucchesi JC, Khanna KK, Ludwig T, Pandita TK. 2005. Involvement of human MOF in ATM function. Mol Cell Biol 25:5292–5305. 10.1128/MCB.25.12.5292-5305.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yang XJ, Ullah M. 2007. MOZ and MORF, two large MYSTic HATs in normal and cancer stem cells. Oncogene 26:5408–5419. 10.1038/sj.onc.1210609. [DOI] [PubMed] [Google Scholar]
- 99.Nielsen OS, Horsman M, Overgaard J. 2001. A future for hyperthermia in cancer treatment? Eur J Cancer 37:1587–1589. 10.1016/s0959-8049(01)00193-9. [DOI] [PubMed] [Google Scholar]
- 100.Pandita TK, Geard CR. 1996. Chromosome aberrations in human fibroblasts induced by monoenergetic neutrons. I. Relative biological effectiveness. Radiat Res 145:730–739. 10.2307/3579364. [DOI] [PubMed] [Google Scholar]
- 101.Anderson RM, Stevens DL, Goodhead DT. 2002. M-FISH analysis shows that complex chromosome aberrations induced by alpha-particle tracks are cumulative products of localized rearrangements. Proc Natl Acad Sci USA 99:12167–12172. 10.1073/pnas.182426799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Kalinich JF, Catravas GN, Snyder SL. 1989. The effect of gamma radiation on DNA methylation. Radiat Res 117:185–197. 10.2307/3577319. [DOI] [PubMed] [Google Scholar]
- 103.Antwih DA, Gabbara KM, Lancaster WD, Ruden DM, Zielske SP. 2013. Radiation-induced epigenetic DNA methylation modification of radiation-response pathways. Epigenetics 8:839–848. 10.4161/epi.25498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Loree J, Koturbash I, Kutanzi K, Baker M, Pogribny I, Kovalchuk O. 2006. Radiation-induced molecular changes in rat mammary tissue: possible implications for radiation-induced carcinogenesis. Int J Radiat Biol 82:805–815. 10.1080/09553000600960027. [DOI] [PubMed] [Google Scholar]
- 105.Pogribny I, Raiche J, Slovack M, Kovalchuk O. 2004. Dose dependence, sex and tissue specificity, and persistence of radiation-induced genomic DNA methylation changes. Biochem Biophys Res Commun 320:1253–1261. 10.1016/j.bbrc.2004.06.081. [DOI] [PubMed] [Google Scholar]
- 106.Kovalchuk O, Burke P, Besplug J, Slovack M, Filkowski J, Pogribny I. 2004. Methylation changes in muscle and liver tissues of male and female mice exposed to acute and chronic low-dose X-ray irradiation. Mutat Res 548:75–84. 10.1016/j.mrfmmm.2003.12.016. [DOI] [PubMed] [Google Scholar]
- 107.Koturbash I, Pogribny I, Kovalchuk O. 2005. Stable loss of global DNA methylation in the radiation-target tissue: a possible mechanism contributing to radiation carcinogenesis? Biochem Biophys Res Commun 337:526–533. 10.1016/j.bbrc.2005.09.084. [DOI] [PubMed] [Google Scholar]
- 108.Koturbash I, Kutanzi K, Hendrickson K, Rodriguez-Juarez R, Kogosov D, Kovalchuk O. 2008. Radiation-induced bystander effects in vivo are sex specific. Mutat Res 642:28–36. 10.1016/j.mrfmmm.2008.04.002. [DOI] [PubMed] [Google Scholar]
- 109.Koturbash I. 2017. LINE-1 in response to exposure to ionizing radiation. Mob Genet Elements 7:e1393491. 10.1080/2159256X.2017.1393491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Luzhna L, Ilnytskyy Y, Kovalchuk O. 2015. Mobilization of LINE-1 in irradiated mammary gland tissue may potentially contribute to low dose radiation-induced genomic instability. Genes Cancer 6:71–81. 10.18632/genesandcancer.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tanaka A, Nakatani Y, Hamada N, Jinno-Oue A, Shimizu N, Wada S, Funayama T, Mori T, Islam S, Hoque SA, Shinagawa M, Ohtsuki T, Kobayashi Y, Hoshino H. 2012. Ionizing irradiation alters the dynamics of human long interspersed nuclear elements 1 (LINE1) retrotransposon. Mutagenesis 27:599–607. 10.1093/mutage/ges025. [DOI] [PubMed] [Google Scholar]
- 112.Farkash EA, Kao GD, Horman SR, Prak ET. 2006. Gamma radiation increases endonuclease-dependent L1 retrotransposition in a cultured cell assay. Nucleic Acids Res 34:1196–1204. 10.1093/nar/gkj522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. 1998. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem 273:5858–5868. 10.1074/jbc.273.10.5858. [DOI] [PubMed] [Google Scholar]
- 114.Shroff R, Arbel-Eden A, Pilch D, Ira G, Bonner WM, Petrini JH, Haber JE, Lichten M. 2004. Distribution and dynamics of chromatin modification induced by a defined DNA double-strand break. Curr Biol 14:1703–1711. 10.1016/j.cub.2004.09.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Fernandez-Capetillo O, Lee A, Nussenzweig M, Nussenzweig A. 2004. H2AX: the histone guardian of the genome. DNA Repair (Amst) 3:959–967. 10.1016/j.dnarep.2004.03.024. [DOI] [PubMed] [Google Scholar]
- 116.Redon C, Pilch DR, Rogakou EP, Orr AH, Lowndes NF, Bonner WM. 2003. Yeast histone 2A serine 129 is essential for the efficient repair of checkpoint-blind DNA damage. EMBO Rep 4:678–684. 10.1038/sj.embor.embor871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Pandita TK, Lieberman HB, Lim DS, Dhar S, Zheng W, Taya Y, Kastan MB. 2000. Ionizing radiation activates the ATM kinase throughout the cell cycle. Oncogene 19:1386–1391. 10.1038/sj.onc.1203444. [DOI] [PubMed] [Google Scholar]
- 118.Mah LJ, El-Osta A, Karagiannis TC. 2010. γH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 24:679–686. 10.1038/leu.2010.6. [DOI] [PubMed] [Google Scholar]
- 119.Fernandez-Capetillo O, Allis CD, Nussenzweig A. 2004. Phosphorylation of histone H2B at DNA double-strand breaks. J Exp Med 199:1671–1677. 10.1084/jem.20032247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.He Z, Cho YY, Ma WY, Choi HS, Bode AM, Dong Z. 2005. Regulation of ultraviolet B-induced phosphorylation of histone H3 at serine 10 by Fyn kinase. J Biol Chem 280:2446–2454. 10.1074/jbc.M402053200. [DOI] [PubMed] [Google Scholar]
- 121.Zhong SP, Ma WY, Dong Z. 2000. ERKs and p38 kinases mediate ultraviolet B-induced phosphorylation of histone H3 at serine 10. J Biol Chem 275:20980–20984. 10.1074/jbc.M909934199. [DOI] [PubMed] [Google Scholar]
- 122.Freyer GA, Palmer DA, Yu Y, Miller RC, Pandita TK. 1996. Neoplastic transformation of mouse C3H10T1/2 cells following exposure to neutrons does not involve mutation of ras gene as analyzed by SSCP and cycle sequencing. Mutat Res 357:237–244. 10.1016/0027-5107(96)00130-3. [DOI] [PubMed] [Google Scholar]
- 123.Hausmann M, Wagner E, Lee J-H, Schrock G, Schaufler W, Krufczik M, Papenfuss F, Port M, Bestvater F, Scherthan H. 2018. Super-resolution localization microscopy of radiation-induced histone H2AX-phosphorylation in relation to H3K9-trimethylation in HeLa cells. Nanoscale 10:4320–4331. 10.1039/c7nr08145f. [DOI] [PubMed] [Google Scholar]
- 124.Simon JA, Kingston RE. 2009. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol 10:697–708. 10.1038/nrm2763. [DOI] [PubMed] [Google Scholar]
- 125.Rath BH, Waung I, Camphausen K, Tofilon PJ. 2018. Inhibition of the histone H3K27 demethylase UTX enhances tumor cell radiosensitivity. Mol Cancer Ther 17:1070–1078. 10.1158/1535-7163.MCT-17-1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Di Nisio E, Lupo G, Licursi V, Negri R. 2021. The role of histone lysine methylation in the response of mammalian cells to ionizing radiation. Front Genet 12:639602. 10.3389/fgene.2021.639602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang Y, Wang Q, Chen S, Hu Y, Yu C, Liu R, Wang Z. 2020. Screening of long noncoding RNAs induced by radiation using microarray. Dose Response 18:1559325820916304. 10.1177/1559325820916304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xiu D, Liu L, Cheng M, Sun X, Ma X. 2020. Knockdown of lncRNA TUG1 enhances radiosensitivity of prostate cancer via the TUG1/miR-139-5p/SMC1A axis. Onco Targets Ther 13:2319–2331. 10.2147/OTT.S236860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Jiang H, Hu X, Zhang H, Li W. 2017. Down-regulation of LncRNA TUG1 enhances radiosensitivity in bladder cancer via suppressing HMGB1 expression. Radiat Oncol 12:65. 10.1186/s13014-017-0802-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Hu X, Ding D, Zhang J, Cui J. 2019. Knockdown of lncRNA HOTAIR sensitizes breast cancer cells to ionizing radiation through activating miR-218. Biosci Rep 39:BSR20181038. 10.1042/BSR20181038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhang S, Wang B, Xiao H, Dong J, Li Y, Zhu C, Jin Y, Li H, Cui M, Fan S. 2020. LncRNA HOTAIR enhances breast cancer radioresistance through facilitating HSPA1A expression via sequestering miR-449b-5p. Thorac Cancer 11:1801–1816. 10.1111/1759-7714.13450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Thapar R, Wang JL, Hammel M, Ye R, Liang K, Sun C, Hnizda A, Liang S, Maw SS, Lee L, Villarreal H, Forrester I, Fang S, Tsai MS, Blundell TL, Davis AJ, Lin C, Lees-Miller SP, Strick TR, Tainer JA. 2020. Mechanism of efficient double-strand break repair by a long noncoding RNA. Nucleic Acids Res 48:10953–10972. 10.1093/nar/gkaa784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Li Z, Zhou Y, Tu B, Bu Y, Liu A, Kong J. 2017. Long noncoding RNA MALAT1 affects the efficacy of radiotherapy for esophageal squamous cell carcinoma by regulating Cks1 expression. J Oral Pathol Med 46:583–590. 10.1111/jop.12538. [DOI] [PubMed] [Google Scholar]
- 134.Lu H, He Y, Lin L, Qi Z, Ma L, Li L, Su Y. 2016. Long noncoding RNA MALAT1 modulates radiosensitivity of HR-HPV+ cervical cancer via sponging miR-145. Tumour Biol 37:1683–1691. 10.1007/s13277-015-3946-5. [DOI] [PubMed] [Google Scholar]
- 135.Zhang H, Luo H, Hu Z, Peng J, Jiang Z, Song T, Wu B, Yue J, Zhou R, Xie R, Chen T, Wu S. 2015. Targeting WISP1 to sensitize esophageal squamous cell carcinoma to irradiation. Oncotarget 6:6218–6234. 10.18632/oncotarget.3358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Gao J, Liu L, Li G, Cai M, Tan C, Han X, Han L. 2019. lncRNA GAS5 confers the radio sensitivity of cervical cancer cells via regulating miR-106b/IER3 axis. Int J Biol Macromol 126:994–1001. 10.1016/j.ijbiomac.2018.12.176. [DOI] [PubMed] [Google Scholar]
- 137.Zhang N, Zeng X, Sun C, Guo H, Wang T, Wei L, Zhang Y, Zhao J, Ma X. 2019. LncRNA LINC00963 promotes tumorigenesis and radioresistance in breast cancer by sponging miR-324-3p and inducing ACK1 expression. Mol Ther Nucleic Acids 18:871–881. 10.1016/j.omtn.2019.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 138.Han D, Wang J, Cheng G. 2018. LncRNA NEAT1 enhances the radioresistance of cervical cancer via miR-193b-3p/CCND1 axis. Oncotarget 9:2395–2409. 10.18632/oncotarget.23416. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 139.Małachowska B, Tomasik B, Stawiski K, Kulkarni S, Guha C, Chowdhury D, Fendler W. 2020. Circulating microRNAs as biomarkers of radiation exposure: a systematic review and meta-analysis. Int J Radiat Oncol Biol Phys 106:390–402. 10.1016/j.ijrobp.2019.10.028. [DOI] [PubMed] [Google Scholar]
- 140.Weidhaas JB, Babar I, Nallur SM, Trang P, Roush S, Boehm M, Gillespie E, Slack FJ. 2007. MicroRNAs as potential agents to alter resistance to cytotoxic anticancer therapy. Cancer Res 67:11111–11116. 10.1158/0008-5472.CAN-07-2858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Liu Z, Liang X, Li X, Liu X, Zhu M, Gu Y, Zhou P. 2019. MiRNA-21 functions in ionizing radiation-induced epithelium-to-mesenchymal transition (EMT) by downregulating PTEN. Toxicol Res (Camb) 8:328–340. 10.1039/c9tx00019d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Banday S, Pandita RK, Mushtaq A, Bacolla A, Mir US, Singh DK, Jan S, Bhat KP, Hunt CR, Rao G, Charaka VK, Tainer JA, Pandita TK, Altaf M. 2021. Autism-associated vigilin depletion impairs DNA damage repair. Mol Cell Biol 41:e0008221. 10.1128/MCB.00082-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Mohammad HP, Baylin SB. 2010. Linking cell signaling and the epigenetic machinery. Nat Biotechnol 28:1033–1038. 10.1038/nbt1010-1033. [DOI] [PubMed] [Google Scholar]
- 144.Castonguay Z, Auger C, Thomas SC, Chahma M, Appanna VD. 2014. Nuclear lactate dehydrogenase modulates histone modification in human hepatocytes. Biochem Biophys Res Commun 454:172–177. 10.1016/j.bbrc.2014.10.071. [DOI] [PubMed] [Google Scholar]
- 145.Zhong XH, Howard BD. 1990. Phosphotyrosine-containing lactate dehydrogenase is restricted to the nuclei of PC12 pheochromocytoma cells. Mol Cell Biol 10:770–776. 10.1128/mcb.10.2.770-776.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Neary CL, Pastorino JG. 2013. Akt inhibition promotes hexokinase 2 redistribution and glucose uptake in cancer cells. J Cell Physiol 228:1943–1948. 10.1002/jcp.24361. [DOI] [PubMed] [Google Scholar]
- 147.Neary CL, Pastorino JG. 2010. Nucleocytoplasmic shuttling of hexokinase II in a cancer cell. Biochem Biophys Res Commun 394:1075–1081. 10.1016/j.bbrc.2010.03.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Gancedo JM. 1998. Yeast carbon catabolite repression. Microbiol Mol Biol Rev 62:334–361. 10.1128/MMBR.62.2.334-361.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Li B, Qiu B, Lee DS, Walton ZE, Ochocki JD, Mathew LK, Mancuso A, Gade TP, Keith B, Nissim I, Simon MC. 2014. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 513:251–255. 10.1038/nature13557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lee SM, Kim JH, Cho EJ, Youn HD. 2009. A nucleocytoplasmic malate dehydrogenase regulates p53 transcriptional activity in response to metabolic stress. Cell Death Differ 16:738–748. 10.1038/cdd.2009.5. [DOI] [PubMed] [Google Scholar]
- 151.Luo W, Hu H, Chang R, Zhong J, Knabel M, O’Meally R, Cole RN, Pandey A, Semenza GL. 2011. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell 145:732–744. 10.1016/j.cell.2011.03.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Li S, Swanson SK, Gogol M, Florens L, Washburn MP, Workman JL, Suganuma T. 2015. Serine and SAM responsive complex SESAME regulates histone modification crosstalk by sensing cellular metabolism. Mol Cell 60:408–421. 10.1016/j.molcel.2015.09.024. [DOI] [PubMed] [Google Scholar]
- 153.Comerford SA, Huang Z, Du X, Wang Y, Cai L, Witkiewicz AK, Walters H, Tantawy MN, Fu A, Manning HC, Horton JD, Hammer RE, McKnight SL, Tu BP. 2014. Acetate dependence of tumors. Cell 159:1591–1602. 10.1016/j.cell.2014.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Sutendra G, Kinnaird A, Dromparis P, Paulin R, Stenson TH, Haromy A, Hashimoto K, Zhang N, Flaim E, Michelakis ED. 2014. A nuclear pyruvate dehydrogenase complex is important for the generation of acetyl-CoA and histone acetylation. Cell 158:84–97. 10.1016/j.cell.2014.04.046. [DOI] [PubMed] [Google Scholar]
- 155.Ariyannur PS, Moffett JR, Madhavarao CN, Arun P, Vishnu N, Jacobowitz DM, Hallows WC, Denu JM, Namboodiri AM. 2010. Nuclear-cytoplasmic localization of acetyl coenzyme a synthetase-1 in the rat brain. J Comp Neurol 518:2952–2977. 10.1002/cne.22373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Matsuda S, Adachi J, Ihara M, Tanuma N, Shima H, Kakizuka A, Ikura M, Ikura T, Matsuda T. 2016. Nuclear pyruvate kinase M2 complex serves as a transcriptional coactivator of arylhydrocarbon receptor. Nucleic Acids Res 44:636–647. 10.1093/nar/gkv967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Chiang PK, Gordon RK, Tal J, Zeng GC, Doctor BP, Pardhasaradhi K, McCann PP. 1996. S-Adenosylmethionine and methylation. FASEB J 10:471–480. 10.1096/fasebj.10.4.8647346. [DOI] [PubMed] [Google Scholar]
- 158.Ye C, Tu BP. 2018. Sink into the epigenome: histones as repositories that influence cellular metabolism. Trends Endocrinol Metab 29:626–637. 10.1016/j.tem.2018.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Sciacovelli M, Frezza C. 2016. Oncometabolites: unconventional triggers of oncogenic signaling cascades. Free Radic Biol Med 100:175–181. 10.1016/j.freeradbiomed.2016.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV, Jr, de Cabo R, Ulrich S, Akassoglou K, Verdin E. 2013. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339:211–214. 10.1126/science.1227166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Rodrigues LM, Uribe-Lewis S, Madhu B, Honess DJ, Stubbs M, Griffiths JR. 2017. The action of beta-hydroxybutyrate on the growth, metabolism and global histone H3 acetylation of spontaneous mouse mammary tumours: evidence of a beta-hydroxybutyrate paradox. Cancer Metab 5:4. 10.1186/s40170-017-0166-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Shukla SK, Gebregiworgis T, Purohit V, Chaika NV, Gunda V, Radhakrishnan P, Mehla K, Pipinos II, Powers R, Yu F, Singh PK. 2014. Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia. Cancer Metab 2:18. 10.1186/2049-3002-2-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, Howell A, Sotgia F, Lisanti MP. 2012. Ketone body utilization drives tumor growth and metastasis. Cell Cycle 11:3964–3971. 10.4161/cc.22137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Lee HG, Kim H, Son T, Jeong Y, Kim SU, Dong SM, Park YN, Lee JD, Lee JM, Park JH. 2016. Regulation of HK2 expression through alterations in CpG methylation of the HK2 promoter during progression of hepatocellular carcinoma. Oncotarget 7:41798–41810. 10.18632/oncotarget.9723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wolf A, Agnihotri S, Munoz D, Guha A. 2011. Developmental profile and regulation of the glycolytic enzyme hexokinase 2 in normal brain and glioblastoma multiforme. Neurobiol Dis 44:84–91. 10.1016/j.nbd.2011.06.007. [DOI] [PubMed] [Google Scholar]
- 166.Goel A, Mathupala SP, Pedersen PL. 2003. Glucose metabolism in cancer. Evidence that demethylation events play a role in activating type II hexokinase gene expression. J Biol Chem 278:15333–15340. 10.1074/jbc.M300608200. [DOI] [PubMed] [Google Scholar]
- 167.Wan W, Peng K, Li M, Qin L, Tong Z, Yan J, Shen B, Yu C. 2017. Histone demethylase JMJD1A promotes urinary bladder cancer progression by enhancing glycolysis through coactivation of hypoxia inducible factor 1α. Oncogene 36:3868–3877. 10.1038/onc.2017.13. [DOI] [PubMed] [Google Scholar]
- 168.Lopez-Serra P, Marcilla M, Villanueva A, Ramos-Fernandez A, Palau A, Leal L, Wahi JE, Setien-Baranda F, Szczesna K, Moutinho C, Martinez-Cardus A, Heyn H, Sandoval J, Puertas S, Vidal A, Sanjuan X, Martinez-Balibrea E, Vinals F, Perales JC, Bramsem JB, Orntoft TF, Andersen CL, Tabernero J, McDermott U, Boxer MB, Vander Heiden MG, Albar JP, Esteller M. 2014. A DERL3-associated defect in the degradation of SLC2A1 mediates the Warburg effect. Nat Commun 5:3608. 10.1038/ncomms4608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Chen M, Zhang J, Li N, Qian Z, Zhu M, Li Q, Zheng J, Wang X, Shi G. 2011. Promoter hypermethylation mediated downregulation of FBP1 in human hepatocellular carcinoma and colon cancer. PLoS One 6:e25564. 10.1371/journal.pone.0025564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mondal P, Sen S, Klein BJ, Tiwary N, Gadad SS, Kutateladze TG, Roy S, Das C. 2020. TCF19 promotes cell proliferation through binding to the histone H3K4me3 mark. Biochemistry 59:389–399. 10.1021/acs.biochem.9b00771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Sen S, Sanyal S, Srivastava DK, Dasgupta D, Roy S, Das C. 2017. Transcription factor 19 interacts with histone 3 lysine 4 trimethylation and controls gluconeogenesis via the nucleosome-remodeling-deacetylase complex. J Biol Chem 292:20362–20378. 10.1074/jbc.M117.786863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Mondal P, Gadad SS, Adhikari S, Ramos EI, Sen S, Prasad P, Das C. 2021. TCF19 and p53 regulate transcription of TIGAR and SCO2 in HCC for mitochondrial energy metabolism and stress adaptation. FASEB J 35:e21814. 10.1096/fj.202002486RR. [DOI] [PubMed] [Google Scholar]
- 173.Chalkiadaki A, Guarente L. 2015. The multifaceted functions of sirtuins in cancer. Nat Rev Cancer 15:608–624. 10.1038/nrc3985. [DOI] [PubMed] [Google Scholar]
- 174.Sebastian C, Zwaans BM, Silberman DM, Gymrek M, Goren A, Zhong L, Ram O, Truelove J, Guimaraes AR, Toiber D, Cosentino C, Greenson JK, MacDonald AI, McGlynn L, Maxwell F, Edwards J, Giacosa S, Guccione E, Weissleder R, Bernstein BE, Regev A, Shiels PG, Lombard DB, Mostoslavsky R. 2012. The histone deacetylase SIRT6 is a tumor suppressor that controls cancer metabolism. Cell 151:1185–1199. 10.1016/j.cell.2012.10.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Hallows WC, Yu W, Denu JM. 2012. Regulation of glycolytic enzyme phosphoglycerate mutase-1 by Sirt1 protein-mediated deacetylation. J Biol Chem 287:3850–3858. 10.1074/jbc.M111.317404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Yang W, Xia Y, Hawke D, Li X, Liang J, Xing D, Aldape K, Hunter T, Alfred Yung WK, Lu Z. 2012. PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell 150:685–696. 10.1016/j.cell.2012.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Chakrabarti A, Chen AW, Varner JD. 2011. A review of the mammalian unfolded protein response. Biotechnol Bioeng 108:2777–2793. 10.1002/bit.23282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Ron D. 2002. Translational control in the endoplasmic reticulum stress response. J Clin Invest 110:1383–1388. 10.1172/JCI16784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Jones CT, Swingler RJ, Simpson SA, Brock DJ. 1995. Superoxide dismutase mutations in an unselected cohort of Scottish amyotrophic lateral sclerosis patients. J Med Genet 32:290–292. 10.1136/jmg.32.4.290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kornfeld R, Kornfeld S. 1985. Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem 54:631–664. 10.1146/annurev.bi.54.070185.003215. [DOI] [PubMed] [Google Scholar]
- 181.Hubbard SC, Ivatt RJ. 1981. Synthesis and processing of asparagine-linked oligosaccharides. Annu Rev Biochem 50:555–583. 10.1146/annurev.bi.50.070181.003011. [DOI] [PubMed] [Google Scholar]
- 182.Schubert U, Anton LC, Gibbs J, Norbury CC, Yewdell JW, Bennink JR. 2000. Rapid degradation of a large fraction of newly synthesized proteins by proteasomes. Nature 404:770–774. 10.1038/35008096. [DOI] [PubMed] [Google Scholar]
- 183.Perri ER, Thomas CJ, Parakh S, Spencer DM, Atkin JD. 2015. The unfolded protein response and the role of protein disulfide isomerase in neurodegeneration. Front Cell Dev Biol 3:80. 10.3389/fcell.2015.00080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Cox JS, Walter P. 1996. A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell 87:391–404. 10.1016/s0092-8674(00)81360-4. [DOI] [PubMed] [Google Scholar]
- 185.Hetz C. 2012. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol 13:89–102. 10.1038/nrm3270. [DOI] [PubMed] [Google Scholar]
- 186.Schroder M, Clark R, Kaufman RJ. 2003. IRE1- and HAC1-independent transcriptional regulation in the unfolded protein response of yeast. Mol Microbiol 49:591–606. 10.1046/j.1365-2958.2003.03585.x. [DOI] [PubMed] [Google Scholar]
- 187.Shen X, Ellis RE, Lee K, Liu CY, Yang K, Solomon A, Yoshida H, Morimoto R, Kurnit DM, Mori K, Kaufman RJ. 2001. Complementary signaling pathways regulate the unfolded protein response and are required for Caenorhabditis elegans development. Cell 107:893–903. 10.1016/s0092-8674(01)00612-2. [DOI] [PubMed] [Google Scholar]
- 188.Nilsen KH. 1976. Malignant lymphoma and rheumatic symptoms. N Z Med J 83:320–322. [PubMed] [Google Scholar]
- 189.Schindler AJ, Schekman R. 2009. In vitro reconstitution of ER-stress induced ATF6 transport in COPII vesicles. Proc Natl Acad Sci USA 106:17775–17780. 10.1073/pnas.0910342106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Shen J, Chen X, Hendershot L, Prywes R. 2002. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of Golgi localization signals. Dev Cell 3:99–111. 10.1016/s1534-5807(02)00203-4. [DOI] [PubMed] [Google Scholar]
- 191.Han D, Lerner AG, Vande Walle L, Upton JP, Xu W, Hagen A, Backes BJ, Oakes SA, Papa FR. 2009. IRE1α kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell 138:562–575. 10.1016/j.cell.2009.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Hollien J, Lin JH, Li H, Stevens N, Walter P, Weissman JS. 2009. Regulated Ire1-dependent decay of messenger RNAs in mammalian cells. J Cell Biol 186:323–331. 10.1083/jcb.200903014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Hollien J, Weissman JS. 2006. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science 313:104–107. 10.1126/science.1129631. [DOI] [PubMed] [Google Scholar]
- 194.Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. 2002. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 415:92–96. 10.1038/415092a. [DOI] [PubMed] [Google Scholar]
- 195.Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, Ron D. 2000. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287:664–666. 10.1126/science.287.5453.664. [DOI] [PubMed] [Google Scholar]
- 196.Acosta-Alvear D, Zhou Y, Blais A, Tsikitis M, Lents NH, Arias C, Lennon CJ, Kluger Y, Dynlacht BD. 2007. XBP1 controls diverse cell type- and condition-specific transcriptional regulatory networks. Mol Cell 27:53–66. 10.1016/j.molcel.2007.06.011. [DOI] [PubMed] [Google Scholar]
- 197.Lee AH, Iwakoshi NN, Glimcher LH. 2003. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 23:7448–7459. 10.1128/MCB.23.21.7448-7459.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, Schapira M, Ron D. 2000. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell 6:1099–1108. 10.1016/s1097-2765(00)00108-8. [DOI] [PubMed] [Google Scholar]
- 199.Ameri K, Harris AL. 2008. Activating transcription factor 4. Int J Biochem Cell Biol 40:14–21. 10.1016/j.biocel.2007.01.020. [DOI] [PubMed] [Google Scholar]
- 200.Tabas I, Ron D. 2011. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol 13:184–190. 10.1038/ncb0311-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Mahameed M, Boukeileh S, Obiedat A, Darawshi O, Dipta P, Rimon A, McLennan G, Fassler R, Reichmann D, Karni R, Preisinger C, Wilhelm T, Huber M, Tirosh B. 2020. Pharmacological induction of selective endoplasmic reticulum retention as a strategy for cancer therapy. Nat Commun 11:1304. 10.1038/s41467-020-15067-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Caretti G, Salsi V, Vecchi C, Imbriano C, Mantovani R. 2003. Dynamic recruitment of NF-Y and histone acetyltransferases on cell-cycle promoters. J Biol Chem 278:30435–30440. 10.1074/jbc.M304606200. [DOI] [PubMed] [Google Scholar]
- 203.Kozlowski L, Garvis S, Bedet C, Palladino F. 2014. The Caenorhabditis elegans HP1 family protein HPL-2 maintains ER homeostasis through the UPR and hormesis. Proc Natl Acad Sci USA 111:5956–5961. 10.1073/pnas.1321698111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Baumeister P, Luo S, Skarnes WC, Sui G, Seto E, Shi Y, Lee AS. 2005. Endoplasmic reticulum stress induction of the Grp78/BiP promoter: activating mechanisms mediated by YY1 and its interactive chromatin modifiers. Mol Cell Biol 25:4529–4540. 10.1128/MCB.25.11.4529-4540.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Schram AW, Baas R, Jansen PW, Riss A, Tora L, Vermeulen M, Timmers HT. 2013. A dual role for SAGA-associated factor 29 (SGF29) in ER stress survival by coordination of both histone H3 acetylation and histone H3 lysine-4 trimethylation. PLoS One 8:e70035. 10.1371/journal.pone.0070035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Wang Y, Alam GN, Ning Y, Visioli F, Dong Z, Nor JE, Polverini PJ. 2012. The unfolded protein response induces the angiogenic switch in human tumor cells through the PERK/ATF4 pathway. Cancer Res 72:5396–5406. 10.1158/0008-5472.CAN-12-0474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Chitnis NS, Pytel D, Bobrovnikova-Marjon E, Pant D, Zheng H, Maas NL, Frederick B, Kushner JA, Chodosh LA, Koumenis C, Fuchs SY, Diehl JA. 2012. miR-211 is a prosurvival microRNA that regulates chop expression in a PERK-dependent manner. Mol Cell 48:353–364. 10.1016/j.molcel.2012.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Updegraff BL, O’Donnell KA. 2013. Stressing the importance of CHOP in liver cancer. PLoS Genet 9:e1004045. 10.1371/journal.pgen.1004045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Moon DO, Park SY, Choi YH, Ahn JS, Kim GY. 2011. Guggulsterone sensitizes hepatoma cells to TRAIL-induced apoptosis through the induction of CHOP-dependent DR5: involvement of ROS-dependent ER-stress. Biochem Pharmacol 82:1641–1650. 10.1016/j.bcp.2011.08.019. [DOI] [PubMed] [Google Scholar]
- 210.Chen X, Iliopoulos D, Zhang Q, Tang Q, Greenblatt MB, Hatziapostolou M, Lim E, Tam WL, Ni M, Chen Y, Mai J, Shen H, Hu DZ, Adoro S, Hu B, Song M, Tan C, Landis MD, Ferrari M, Shin SJ, Brown M, Chang JC, Liu XS, Glimcher LH. 2014. XBP1 promotes triple-negative breast cancer by controlling the HIF-1α pathway. Nature 508:103–107. 10.1038/nature13119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Pluquet O, Dejeans N, Bouchecareilh M, Lhomond S, Pineau R, Higa A, Delugin M, Combe C, Loriot S, Cubel G, Dugot-Senant N, Vital A, Loiseau H, Gosline SJ, Taouji S, Hallett M, Sarkaria JN, Anderson K, Wu W, Rodriguez FJ, Rosenbaum J, Saltel F, Fernandez-Zapico ME, Chevet E. 2013. Posttranscriptional regulation of PER1 underlies the oncogenic function of IREα. Cancer Res 73:4732–4743. 10.1158/0008-5472.CAN-12-3989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Wang Q, Mora-Jensen H, Weniger MA, Perez-Galan P, Wolford C, Hai T, Ron D, Chen W, Trenkle W, Wiestner A, Ye Y. 2009. ERAD inhibitors integrate ER stress with an epigenetic mechanism to activate BH3-only protein NOXA in cancer cells. Proc Natl Acad Sci USA 106:2200–2205. 10.1073/pnas.0807611106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Tubbs JL, Pegg AE, Tainer JA. 2007. DNA binding, nucleotide flipping, and the helix-turn-helix motif in base repair by O6-alkylguanine-DNA alkyltransferase and its implications for cancer chemotherapy. DNA Repair (Amst) 6:1100–1115. 10.1016/j.dnarep.2007.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Thapar R, Bacolla A, Oyeniran C, Brickner JR, Chinnam NB, Mosammaparast N, Tainer JA. 2019. RNA modifications: reversal mechanisms and cancer. Biochemistry 58:312–329. 10.1021/acs.biochem.8b00949. [DOI] [PubMed] [Google Scholar]
- 215.Payne MJ, Pratap SE, Middleton MR. 2005. Temozolomide in the treatment of solid tumours: current results and rationale for dosing/scheduling. Crit Rev Oncol Hematol 53:241–252. 10.1016/j.critrevonc.2004.10.004. [DOI] [PubMed] [Google Scholar]
- 216.Chang SP, Shen SC, Lee WR, Yang LL, Chen YC. 2011. Imatinib mesylate induction of ROS-dependent apoptosis in melanoma B16F0 cells. J Dermatol Sci 62:183–191. 10.1016/j.jdermsci.2011.03.001. [DOI] [PubMed] [Google Scholar]
- 217.Liou GY, Storz P. 2010. Reactive oxygen species in cancer. Free Radic Res 44:479–496. 10.3109/10715761003667554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Oliveira MS, Barbosa MIF, de Souza TB, Moreira DRM, Martins FT, Villarreal W, Machado RP, Doriguetto AC, Soares MBP, Bezerra DP. 2019. A novel platinum complex containing a piplartine derivative exhibits enhanced cytotoxicity, causes oxidative stress and triggers apoptotic cell death by ERK/p38 pathway in human acute promyelocytic leukemia HL-60 cells. Redox Biol 20:182–194. 10.1016/j.redox.2018.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Bauer D, Werth F, Nguyen HA, Kiecker F, Eberle J. 2017. Critical role of reactive oxygen species (ROS) for synergistic enhancement of apoptosis by vemurafenib and the potassium channel inhibitor TRAM-34 in melanoma cells. Cell Death Dis 8:e2594. 10.1038/cddis.2017.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Shan F, Shao Z, Jiang S, Cheng Z. 2016. Erlotinib induces the human non-small-cell lung cancer cells apoptosis via activating ROS-dependent JNK pathways. Cancer Med 5:3166–3175. 10.1002/cam4.881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Alas S, Ng CP, Bonavida B. 2002. Rituximab modifies the cisplatin-mitochondrial signaling pathway, resulting in apoptosis in cisplatin-resistant non-Hodgkin’s lymphoma. Clin Cancer Res 8:836–845. [PubMed] [Google Scholar]
- 222.Renschler MF. 2004. The emerging role of reactive oxygen species in cancer therapy. Eur J Cancer 40:1934–1940. 10.1016/j.ejca.2004.02.031. [DOI] [PubMed] [Google Scholar]
- 223.Kotamraju S, Chitambar CR, Kalivendi SV, Joseph J, Kalyanaraman B. 2002. Transferrin receptor-dependent iron uptake is responsible for doxorubicin-mediated apoptosis in endothelial cells: role of oxidant-induced iron signaling in apoptosis. J Biol Chem 277:17179–17187. 10.1074/jbc.M111604200. [DOI] [PubMed] [Google Scholar]
- 224.Hwang PM, Bunz F, Yu J, Rago C, Chan TA, Murphy MP, Kelso GF, Smith RA, Kinzler KW, Vogelstein B. 2001. Ferredoxin reductase affects p53-dependent, 5-fluorouracil-induced apoptosis in colorectal cancer cells. Nat Med 7:1111–1117. 10.1038/nm1001-1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, Kyle S, Meuth M, Curtin NJ, Helleday T. 2005. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434:913–917. 10.1038/nature03443. [DOI] [PubMed] [Google Scholar]
- 226.Houl JH, Ye Z, Brosey CA, Balapiti-Modarage LPF, Namjoshi S, Bacolla A, Laverty D, Walker BL, Pourfarjam Y, Warden LS, Babu Chinnam N, Moiani D, Stegeman RA, Chen MK, Hung MC, Nagel ZD, Ellenberger T, Kim IK, Jones DE, Ahmed Z, Tainer JA. 2019. Selective small molecule PARG inhibitor causes replication fork stalling and cancer cell death. Nat Commun 10:5654. 10.1038/s41467-019-13508-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Brosey CA, Ho C, Long WZ, Singh S, Burnett K, Hura GL, Nix JC, Bowman GR, Ellenberger T, Tainer JA. 2016. Defining NADH-driven allostery regulating apoptosis-inducing factor. Structure 24:2067–2079. 10.1016/j.str.2016.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.O’Connor MJ. 2015. Targeting the DNA damage response in cancer. Mol Cell 60:547–560. 10.1016/j.molcel.2015.10.040. [DOI] [PubMed] [Google Scholar]
- 229.Hambarde S, Tsai CL, Pandita RK, Bacolla A, Maitra A, Charaka V, Hunt CR, Kumar R, Limbo O, Le Meur R, Chazin WJ, Tsutakawa SE, Russell P, Schlacher K, Pandita TK, Tainer JA. 2021. EXO5-DNA structure and BLM interactions direct DNA resection critical for ATR-dependent replication restart. Mol Cell 81:2989–3006. 10.1016/j.molcel.2021.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Tsutakawa SE, Tsai CL, Yan C, Bralic A, Chazin WJ, Hamdan SM, Scharer OD, Ivanov I, Tainer JA. 2020. Envisioning how the prototypic molecular machine TFIIH functions in transcription initiation and DNA repair. DNA Repair (Amst) 96:102972. 10.1016/j.dnarep.2020.102972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Vancsik T, Forika G, Balogh A, Kiss E, Krenacs T. 2019. Modulated electro-hyperthermia induced p53 driven apoptosis and cell cycle arrest additively support doxorubicin chemotherapy of colorectal cancer in vitro. Cancer Med 8:4292–4303. 10.1002/cam4.2330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Yang KL, Huang CC, Chi MS, Chiang HC, Wang YS, Hsia CC, Andocs G, Wang HE, Chi KH. 2016. In vitro comparison of conventional hyperthermia and modulated electro-hyperthermia. Oncotarget 7:84082–84092. 10.18632/oncotarget.11444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Ansari I, Chaturvedi A, Chitkara D, Singh S. 2021. CRISPR/Cas mediated epigenome editing for cancer therapy. Semin Cancer Biol 10.1016/j.semcancer.2020.12.018. [DOI] [PubMed] [Google Scholar]
- 234.Pulecio J, Verma N, Mejia-Ramirez E, Huangfu D, Raya A. 2017. CRISPR/Cas9-based engineering of the epigenome. Cell Stem Cell 21:431–447. 10.1016/j.stem.2017.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Ermolaeva MA, Segref A, Dakhovnik A, Ou HL, Schneider JI, Utermohlen O, Hoppe T, Schumacher B. 2013. DNA damage in germ cells induces an innate immune response that triggers systemic stress resistance. Nature 501:416–420. 10.1038/nature12452. [DOI] [PMC free article] [PubMed] [Google Scholar]


