Crosstalk between autophagy and intracellular radiation response (Review)

  • Authors:
    • Lelin Hu
    • Hao Wang
    • Li Huang
    • Yong Zhao
    • Junjie Wang
  • View Affiliations

  • Published online on: October 5, 2016     https://doi.org/10.3892/ijo.2016.3719
  • Pages: 2217-2226
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Autophagy induced by radiation is critical to cell fate decision. Evidence now sheds light on the importance of autophagy induced by cancer radiotherapy. Traditional view considers radiation can directly or indirectly damage DNA which can activate DNA damage the repair signaling pathway, a large number of proteins participating in DNA damage repair signaling pathway such as p53, ATM, PARP1, FOXO3a, mTOR and SIRT1 involved in autophagy regulation. However, emerging recent evidence suggests radiation can also cause injury to extranuclear targets such as plasma membrane, mitochondria and endoplasmic reticulum (ER) and induce accumulation of ceramide, ROS, and Ca2+ concentration which activate many signaling pathways to modulate autophagy. Herein we review the role of autophagy in radiation therapy and the potent intracellular autophagic triggers induced by radiation. We aim to provide a more theoretical basis of radiation-induced autophagy, and provide novel targets for developing cytotoxic drugs to increase radiosensitivity.

1. Introduction

Radiotherapy is an effective strategy for the treatment of many kinds of cancer to kill or control the malignant tumor cells. However, its benefits have been limited due to radiation resistance. It is imperative to identify new targets and develop cytotoxic drugs to increase radiosensitivity. Radiosensitization has traditionally been performed with agents known to induce apoptosis. However, recent studies demonstrate autophagy induced by radiation also plays an important role in cancer cell fate decision, particularly in solid tumors (1). Autophagy is an evolutionarily conserved catabolic process that delivers cellular constituents, including damaged or superfluous organelles and long-lived proteins to lysosomes for degradation and recycling, thereby regulating cellular homeostasis. The key steps of autophagy include initiation, phagophore nucleation and elongation, fusion and degradation. Autophagy initiation begins with the membrane distention from either the ER or Golgi complex, followed by formation of autophagosome, a cup shaped double-membrane structure which sequesters cellular components. Subsequently, autophagosome fuses with the lysosome to form an autolysosome which is the site for degradation of the sequestered cargo by the action of the lysosomal hydrolytic enzymes (2).

More than 40 autophagy-related proteins have been identified by large-scale genetic screening in yeast and various mammalian species (3). The core pathway of mammalian autophagy comprises at least four molecular components, including: i) ULK complex (comprising ULK1, ATG13, ATG101 and FIP200), and plays a key role in the induction of autophagy. Autophagy inducers cause dephosphorylation of ULK1; the ULK1 complex then dissociates from the mTORC1 complex, which leads to increasing activity of the BECN1 complex through phosphorylation of AMBRA1 and BECN1 (Beclin 1, autophagy related); ii) core PtdIns3K complex (comprising PIK3C3, PIK3R4/Vps15 and BECN1); BECN1 is a component of core PtdIns3K complex, which participates in phagophore nucleation and elongation. Phosphorylation BECN1 recruits ATG14, AMBRA1 or UVRAG to the PtdIns3K complexes to promote autophagy (4); iii) ATG9 and VMP1, two transmembrane proteins, are critical in leading to autophagosome formation. The biogenesis of the phagophore is thought to initiate at the phagophore assembly site (PAS). Most of the ATG proteins transiently reside at the PAS, although the ultrastructure of this site and the interactions among the ATG proteins during phagophore formation are not known (5). ULK1 complex assembles at the PAS where autophagy is initiated and recruits ATG9-vesicles in order to initiate the formation of autophagosomes (6). VMP1 induced by autophagy stimulus interacts with the BECN1 BH3 domain partitioning BECN1 to the autophagic pathway. This interaction promotes the recruitment of the autophagy-specific PtdIns3K complex to the PAS and activates the PtdIns3K complex, which generates the PtdIns3p. PtdIns3p is necessary to recruit the rest of the ATG machinery, leading to autophagosome formation (7); iv) ATG12 and ATG 8/LC3, are two ubiquitin-like protein conjugation systems essential for autophagosome formation. Ubiquitin-like molecule ATG12 is activated and transferred to the E2-like conjugating enzyme ATG10 by E1-like enzyme ATG7, ultimately attached to ATG5 4, 6, 7. In addition, ATG7 and the E2-like enzyme ATG3 promote the LC3 (ATG8 in yeast) is conjugated to the lipid phosphatidylethanolamine (8). Elongation of the phagophore membrane is dependent on the ATG12 and LC3 conjugation systems (9). Most of these core autophagy pathway components are directly controlled by cellular stress signals (10,11).

Autophagy induced by radiation play bi-directional effects in cell fate decision whether cells survive or die depends on severity and duration of this phenomenon (12). When the stress is mild, autophagy can degrade and recycle damaged or unwanted cellular constituents in autophagolysosomal vesicles to provide additional energy supply during stress, which has an essential effect in quality control of organelles and cellular adaptation to stress (13). However, several reports demonstrate that various drugs in combination with radiation promote autophagic cell death rather than apoptosis significantly contributing to the antitumor effects of radiosensitizing treatment or radiotherapy in glioma (14). Hence, autophagy induced by radiation also functions as a pro-death mechanism. Besides dual activity of autophagy on tumor cell fate in vitro, recently in vivo studies demonstrate that irradiation-induced autophagy exerts a crucial activity on tumor clearance by the immune system. Autophagy induced by radiation contributes to the release of cell death-associated danger signals ATP and HMGB1 that trigger antitumor host immune responses. Furthermore, autophagy inhibition reduces radioresponses in vivo due to deficient immunogenic signaling (15,16).

The initiation signal of autophagy induced by radiation have not been completely elucidated, although biochemical analysis performed during the last few years has identified several proteins in DNA damage repair signaling pathway or oxidative stress signaling pathway participated in modulation of autophagy. Clarifying crosstalk between autophagy and intracellular radiation response is beneficial to provide novel targets for exploiting novel radiosensitizers to enhance the effects of anticancer therapies for cancer patient.

2. The role of autophagy in radiotherapy

Function of autophagy on tumor cell fate in vitro

A large amount of recent studies have shown that tumor resistance to radiation therapy is often associated with the upregulation of autophagy in many kinds of tumor cell lines, such as colon cancer cells, prostate cancer cells, malignant glioma cells, nasopharyngeal carcinoma cells, and breast cancer cells (17,18). The putative cytoprotective function of autophagy induced by radiation is generally considered to reflect the capacity of the cell to eliminate toxic species such as free radicals and damaged and unwanted proteins or organelles to generate energy and metabolic precursors (19). Due to the cytoprotective function of autophagy, autophagy inhibitors (such as chloroquine, bafilomycin, 3-methyl adenine, or ammonium chloride) and genetic silencing or knockdown of autophagy-associated genes (such as BECN1, ATG 5, 7 or 12) have the potential to be exploited to increase tumor cell radiosensitivity, usually via the promotion of apoptosis (15).

Moreover, some studies reported that autophagy enhanced the anticancer effects of radiotherapy on patients with oral squamous cell carcinoma and glioblastoma cells (20,21). Vitamin D and its analogs such as EB 1089 could enhance the response to radiation in breast cancer and non-small cell lung cancer through the promotion of a cytotoxic form of autophagy (22,23). Hence, autophagy is also recognized as having the potential to contribute to cell killing in response to radiation. This kind of cell death was defined as autophagic cell death or type II apoptosis (24). Autophagic cell death can serve as a part of backup cell death form when stress-induced apoptosis is blocked with broad spectrum caspase inhibitor, z-VAD-Fmk, autophagy can provide an alternative cell death mechanism (25). Moreover, autophagy is important to the radiation-induced senescence. Inhibition of autophagy results in a switch from radiation-induced senescence to apoptosis in breast cancer cells (26).

Function of autophagy on tumor cell fate in vivo

Besides dual activity of autophagy on tumor cell fate in vitro, recent in vivo studies showed autophagy play a crucial role in immunogenic cell death (ICD) induced by radiation (27). After exposure to radiation, autophagy contributes to the release of cell deathassociated danger signals such as calreticulin exposure and HMGB1 and ATP release from dying cells which in turn is required for attracting immune cells including dedritic protein into the tumor that trigger antitumor host immune responses, the molecular machinery that underlies this crucial manifestation of ICD has not yet been elucidated in detail (16,28). The ATP release from dying cells depends on autophagy (29). Recent evidence shows that blocking the autophagic machinery with small interfering RNA prevents the release of the immune stimulating ‘danger signal’, ATP, in chemotherapy treated tumor cells undergoing ICD (30). Combining radiotherapy with specific chemotherapies or immunotherapies is able to induce a repertoire of cancer specific immunogens, which not only potentiate tumor control by enhancing cell kill, but also through the induction of a successful antitumor vaccination. However, the release of ATP may be just a part of an immune stimulating process after exposure to radiation (27). Chloroquine, an autophagy inhibitor which can reduce the release of ATP, still can improve the efficacy of breast cancer radiation therapy by blocking endosomal pathways, which promote an immunogenic form of cell death and better antigen cross-presentation to increase intrinsic radiosensitivity (31). An overview of the effect of autophagy induced by radiation in vivo and in vitro described in this section is presented in Fig. 1.

3. DNA damage repair and autophagy

ATM and autophagy

Radiation injury to DNA is caused directly or indirectly and is known to be repaired immediately (32). ATM, a primary sensor of DNA damage after exposure to radiation, plays a crucial role in initiating autophagy (33). In response to genotoxic stress, DNA damage is detected by MNR (comprising MRE11, NBS1 and Rad50 proteins) and RPA (human replication protein A) complexes which act as sensors and recruit ATM and ATR (ataxia-telangiectasia and RAD3 related) to the site of the lesion, these kinases are activated by DNA lesions and direct the DDR pathways through phosphorylation of downstream targets (34). The activation of ATM/ATR triggers phosphorylation of its downstream targets such as p53, mTOR and FOXO3a, which could trigger radiation-induced autophagy (35).

p53 and autophagy

p53, a critical component of the cellular reaction to radiation, integrates DNA damage signals and radiation-responsive autophagy (36). In unstressed cells, p53 is maintained at low levels by the action of MDM2, an oncogenic E3 ligase, which is essential for both degradation and nuclear export of p53. Following irradiation p53 tends to be rapidly stabilized by reversible post-translational modifications (37). The function of p53 in response to radiation is ATM-dependent. Activation of ATM results from the DNA DSBs, which in turn phosphorylates p53 at Ser15 and Ser20. Whereas, p53 and MDM2 are phosphorylated by ATM directly at Ser395. Both phosphorylated p53 at Ser15 and Ser20 and phosphorylated MDM2 interrupt the binding of MDM2 with p53, and protect p53 from ubiquitination and degradation. At the same time p53 levels are increased, and majority of p53 translocates to the nucleus. As an important transfection factor, p53 promotes autophagy by transactivating its target genes that involved the diversity of radiation responsive pathways in mammalian cells (38). One mechanism for p53-induced autophagy is through activation of energy sensor AMPK pathway and inhibition of the mTOR pathway. Another mechanism of p53-induced autophagy is mediated by transactivating multiple genes with proautophagic roles, including DRAM, PTEN (an inhibitor of the PI3K/AKT signaling pathway), IGF-BP3, DAPK-1 ARF, JNK, Sestrin1/2, IGF-BP3 and PUMA (3941). Recently, Cui et al, verified radiation induced autophagic cell death via the p53/DRAM signaling pathway in breast cancer cells (42).

mTOR and autophagy

mTOR, an atypical serine/threonine kinase, can modify downstream molecules and inhibits autophagy. As an evolutionarily conserved kinase, mTOR functions largely as the catalytic subunit of two distinct protein kinase complexes the mTORC1 (comprising mTOR, mLST8 and Raptor) which is a rapamycin sensitive complex (mTORC1) and the mTORC2 (comprising mTOR, mLST8 and Rictor), which is a rapamycin insensitive complex. Raptor and Rictor serve as scaffolding proteins to regulate the assembly, localization and substrate binding of mTORC1 and mTORC2, respectively (43). Previous studies have reported that altered mTORC1 signaling pathways were mostly found in many human cancers, whereas mTORC2 was less correlated (44). Once mTORC1 is activated in DNA damage repair signaling pathway, mTORC1-mediated phosphorylation prevents activation of ULK1 and its interacting partner ATG13 as well as AMBRA1, the key link between the ULK1 and BECN1 complex. In addition, the activation of mTORC1 prevents AMPK-mediated phosphorylation which is required for full activation of ULK1, which is a serine/threonine kinase that plays a key role in initiator of autophagosome formation in mammalian systems (45,46). The blockade of the mTORC1 can activate the ULK1 complex and promote the interaction with the PtdIns3K complex which is necessary for the formation of new autophagosomes (47). Several studies have reported on the positive effects of mTOR inhibitor on sensitizing cancers to radiation therapy and chemotherapy in lung cancer, pancreatic carcinoma cells and esophageal carcinoma cells (4851).

FOXO3a and autophagy

Under normal conditions, FOXO3a is attached to DNA. The carboxyl-terminal domain (amino acids 616–623) of FOXO3a binds to the FAT domain of ATM, (amino acids 1960–2566), which has been postulated as a protein-binding domain, activation has been shown to depend on its autophosphorylation at Ser 1981 contained in the FAT domain (52). In response to DNA damage induced by radiation the transcriptional activity and protein expression level of FOXO3a increase. Furthermore, radiation can induce the nuclear translocation of FOXO3a in Saos2 cells (53). FOXO3a localization within the nuclear phospho-ATM (Ser1981) foci in irradiated cells is affected by its posttranslational modification (phospho-Thr32) (54). Nuclear transcription factor FOXO3a detaches from DNA to interact with ATM and activated ATM via phosphorylation, which can trigger autophagy as describe above. Additionally, FOXO3a regulates transcription of autophagy-related genes, including LC3 or BNIP3 (55).

PARP-1 and autophagy

PARP-1 is another protein of DDR involved in the regulation of autophagy. PARP-1 is a member of a family of 18 such proteins which are known to bind to DNA and function in DNA damage repair. Irradiation is known to induce DNA damage and activate PARP-1. The important role of PARP-1 in radiation response and the efficacy of PARP-1 inhibitors as radiosensitizers have been investigated for more than 30 years (56). Following DNA binding, The PARP-1 enzymatic activity is triggered caused by depletion of ATP and activation of AMPK. The PARP-1 activation can not only lead to the inhibition of mTOR which induces autophagy but also promotes HMGB1-mediated autophagy (57,58). Chen, et al reported that radiation activates PARP-1 which regulates autophagy via the activation of AMPK or inhibition mTOR in CNE-2 human nasopha ryngeal carcinoma cells (59,60).

SIRT1 and autophagy

SIRT1, a member of the mammalian sirtuin family, plays an important role in autophagy. The mechanism of SIRT1 in regulating this process is due to its ability to deacetylate histones and non-histone proteins, such as p53, FOXO3a and H4K16 (lysine 16 on histone H4) in response to stresses and DNA damage (61). SIRT1 interacts with p53 and affects the transcriptional activity of p53 which can be, directly or indirectly, involved in DDR. SIRT1 regulates autophagy by both epigenetic and posttranslational mechanisms (62). SIRT1 regulates autophagy gene H4K16 (lysine 16 on histone H4) expression through histone deacetylation. H4K16 deacetylation inhibits the transcription of genes involved in the early and late steps of autophagy in multiple cell types, resulting in decreased autophagic flux. Moreover, SIRT1 indirectly regulates autophagy by deacetylation of FOXO3a, leading to increased expression of autophagy-related genes, including BNIP3, which are critical for autophagy induction.

4. Mitochondrial damage and autophagy

ATP and autophagy

Growing evidence has suggested that mitochondria may also be an important extranuclear target in mediating the cytotoxic effects of radiation (63,64). Mitochondria are essential organelles which perform multiple functions. During the process of tricarboxylic acid cycle and oxidative phosphorylation, mitochondria utilize oxygen to generate ATP from organic fuel molecules using the electrochemical gradient generated across the inner of two membranes by the electron transport chain, but in the process also produce ROS (65). Radiation promote the accumulation of ROS, which acts straightforwardly by direct modifications of biological molecules like proteins, lipids and nucleic acids and leads to mitochondrial dysfunction post-irradiation and causes an energy imbalance.

Once the AMP/ATP ratio increases, AMPK, a genuine sensor of the energetic state of the cell, was activated. AMPK which directly responds to the so-called adenylate energy charge as the enzyme is activated by very low increases of AMP levels (and, to certain extent, of ADP), and deactivated by ATP, and could be activated by upstream kinases. Phospho-active AMPK activates autophagy to restore the correct adenylate energy charge. At the molecular level, active AMPK stimulates autophagy by means of at least four distinct mechanisms. These include: i) phosphorylation of the mTORC1 inhibitor, TSC2 at Ser1387, which induces RHEB GTPase activity; RHEB-GDP strongly inhibits the activity of mTOR and activate autophagy, and does so both in vitro and in vivo (66); ii) phosphorylation of ULK1 at Ser317 and Ser777. ULK1 is then free to interact with and to be phosphorylated by AMPK (67); iii) AMPK regulates autophagy by phosphorylating PIK3C3 and BECN1 at Thr388, S90 and S93; iv) AMPK regulates autophagy not only by promoting formation of the PIK3C3-BECN1 complex, but also by inhibiting the interaction of BECN1 and BCL2 to release more available BECN1 to form PtdIns3K complexes (68).

Mitochondrial ROS and mitophagy

Mitochondria are one of target organelles for low-dose radiation (69). Recently, Shimura et al reported that low-dose, long-term fractionated radiation induces oxidative stress in irradiated normal human fibroblasts via accumulation of mitochondrial ROS (70). Mitochondria ROS act as essential signaling molecules in the regulation of the mitophagy and tumor development (71,72). Mitophagy is a process of mitochondria-selective autophagy in response to various signals, including oxidative stress, starvation, and modification of mitochondrial proteins (73). It has been shown that degradation of mitochondria and mtDNA can be executed by mitophagy. Mitophagy is mediated by two different molecular pathways: BNIP3/NIX and Parkin (PARK2)/PINK1 (PTEN induced putative kinase 1). BNIP3L/NIX, localizes to mitochondria, and regulates the aggregation of damaged mitochondria to autophagosomes by directly interacting with LC3 or GABARAP (GABA[A] receptor-associated protein). 125I-irradiation elevated the expression of BNIP3 (69). BNIP3 is one of the primary death promoting proteins, which competes with BECN1 for binding to BCL2, and hence releases ‘free’ BECN1 to induce autophagy. Additionally, radiation increased mitochondrial mass and caused accumulation of mitochondrial ROS, which resulted in mitochondrial damage that was in turn recognized by Parkin. Parkin/PINK1 allows the selective degradation of damaged and dysfunctional mitochondria in response to mitochondrial membrane de-polarization induced by ROS. Parkin accumulated in depolarized fragmented mitochondria by the recruitment of PINK1 to control mitochondrial quality via induction of mitophagy (69,74). As an E3 ubiquitin ligase, the ligase activity of Parkin was activated by PINK1. This process involves the ubiquitination of VDAC1 (voltage-dependent anion channel 1) and its combination with p62, which subsequently interacts with LC3 and directs this complex to the autophagosome (75).

5. ER stress and autophagy

As previously indicated ER is another target of radiation, radiation initially induces ROS production, which would activate ER membrane sensors of ER stress in IEC-6 cells and breast cancer MDA-MB-231 and MCF-7 cells (76,77). ER stress induced by radiation triggers signal transduction pathways, known as unfolded protein response (UPR) also through the induction of DNA damage (78,79). ER stress and UPR related genes are required for stress induced autophagy. The last few years have evidenced that the ER stress and autophagy processes are closely related as some of the signaling routes activated during the ER stress response are involved in stimulating autophagy.

Although the main signaling pathway of ER stress that is activated following irradiation is still a debatable one; some recent evidence suggests that PERK-eIF2a and/or IRE1a may serve as the main executing pathways of ER stress in irradiation scenarios (8082). A link between autophagy and the ER stress has been further substantiated by the PERK-eIF2α pathway which is essential for autophagy induction after ER stress. PERK phosphorylates the eukaryotic initiation factor 2α (eIF2α) on residue serine 51, which then initiates a cascade of events that decreases the overload of misfolded proteins, thereby alleviating ER stress (83). eIF2α phosphorylation stimulates the selective translation of the ATF4 transcription factor (although general translation is shut off) and CHOP (a transcription factor induced by ATF4), which were shown to transcriptionally regulate more than a dozen ATG genes. These genes are necessary for sustained autophagy (84). PERK activated by oxidative stress can also directly inhibit mTOR or indirectly inhibits mTOR via activation of PARP1, which can induce autophagy (85). In addition, IRE1 was activated in response to a variety of cellular stressors after exposure to radiation. In mammalian cells, IRE1-JNK pathway was required for autophagy activation after ER stress (86,87). Activation of IRE1 and JNK causes Bcl-2 phosphorylation, dissociation of BECN1, activation of the PI3K complex and induces autophagy. Moreover, dysregulated autophagy may also trigger the IRE1 activity with concomitant activation of UPR, thereby dampening excessive autophagy triggered via the PERK/eIF2α pathway and pointing to a plausible feedback mechanism in the control of UPR signaling (88).

6. ROS and autophagy

ROS is a family of highly reactive molecules which includes free oxygen radicals, like superoxide anion (O2•−), hydroxyl radical (OH), and non-radical oxygen derivatives, like the stable hydrogen peroxide (H2O2). ROS are unstable molecules which have the capacity to readily convert to many different viable and active forms. The superoxide radicals react to form other ROS, namely, hydrogen peroxides and hydroxyl radicals, and interconvert with RNS, which generate effects similar to ROS (89). Radiation exposure has been intimately linked to increased reactive ROS production and persistent oxidative stress in cells. Accumulating data implicate accumulation of ROS from radiolysis of water molecules, damage mitochondria and ER regulation of autophagy in oxidative stress response (66,90). Although multiple studies reported that accumulation of ROS regulated autophagy by pathways such as: i) oxidization of ATG4 leading to accumulation of autophagosomes; ii) activation of the AMPK signaling cascade inducing the initiation of autophagy through the ULK1 complex; iii) disruption of BECN1-BCL-2 interaction leading to the initiation of autophagy; or iv) alteration of mitochondria homeostasis leading to mitophagy activation (75). Furthermore, reactive oxygen/nitrogen species (ROS/RNS) generated in the context of radiation exposure are essential activators of cytoplasmic signaling cascades such as p38 MAPK, JNK, HIF-1α, which play essential roles in the regulation of autophagy (91).

p38 MAPK and JNK are two of the major MAPK subfamilies, which are serine/threonine kinases that mediate responses to various extracellular stimuli. Once activated in response to various stresses, p38 MAPK and JNK lead to phosphorylation-dependent activation of other kinases and transcription factors and play a crucial role in the modulation of autophagy (92). It has been previously reported that disruption of ROS clearance activates the p38 MAPK pathway which acts as a key regulator in ROS-mediated autophagy (93). The activation of p38 MAPK was in part responsible for the downregulation of phosphorylated mTOR and the subsequent activation of autophagy (94). p38 MAPK depletion is also found to regulate the trafficking of ATG9, a multi-spanning membrane protein that is essential for autophagy (95). Fractionated radiation activated p38 MAPK which is involved in radiation-induced stabilization of HIF-1α which play an important role in switching autophagy. Consistently, inhibition of p38 MAPK also effectively attenuated radiation-induced stabilization of HIF-1α (96).

After exposure to radiation, the expression of HIF-1α increased, then HIF-1α translocated into the nucleus. In the nucleus, HIF-1α bound to the hypoxia response element in its target promoter and promoted the transcription of its target (97). HIF-1α is a subunit of HIF1 which consists of a regulatory HIF-1α subunit and the constitutively expressed HIF-1β subunit, both are members of the basic helix-loop-helix and PER-ARNT-SIM families of transcription factors (98). Pervious opinion considered that while HIF-1β is constitutively expressed, HIF-1α is an oxygen sensitive subunit and its expression is induced under hypoxic conditions (99). Accumulating evidence has recently revealed that HIF-1α is activated in cancer cells not only under hypoxic conditions, but also in the presence of oxygen when the following conditions are satisfied. In addition to post-translational mechanisms, regulation at the level of transcription and translation initiation is also important for the activation of HIF-1α under normoxic conditions (100). Koshikawa et al reported that ROS generated in mitochondria upregulated the transcription of the HIF-1α gene via the PI3K-Akt/PKC/ HDAC pathway, leading to the accumulation and activation of HIF-1α in tumor cells (101). ROS upregulates HIF-1α transcription by activating non-hypoxic factors in a redox-sensitive manner (102). HIF-1α induces autophagy by a mechanism involving the upregulated expression of its target genes BNIP3 and its homologue NIX and the dissociation of the BECN1-BCL2 complex, leading to release of BECN1, which is capable of triggering autophagy (103,104).

7. Ceramide and autophagy

Radiation activates the cell surface glycohydrolases which play a crucial role in the production of ceramide at the plasma membrane. Ceramide is localized within lipid rafts in the plasma membrane, which are specialized membrane microdomains that regulate various signaling pathways (105). Ceramide is also a central molecule of sphingolipid metabolism and involved in the regulation of autophagy at various levels (106,107). Ceramide downregulates the expression of amino acid and nutrient transporters in the context of starvation, a state that induces survival autophagy by reducing mTOR signaling pathway or activating AMPK (108,109). Further evidence indicated that ceramide increases BECN1 expression by activating JNK kinase, which in turn activates c-Jun, a known transcription factor for BECN1 expression (110). Ceramide accumulation also can induce ER stress, mTOR inhibition via TRB3 (tribbles homologue 3), which triggered lethal autophagy (111113). Moreover, ceramide can be generated by neutral sphingomyelinase in mitochondria, which is a late event upon the cell irradiation following the early phase of ceramide accumulation at the cell plasma membrane (106). Ceramide accumulation in the mitochondria can lead to ceramide stress-induced mitochondrial fragmentation, and decrease in ATP production (114). These important mitochondrial parameter changes can induce mitophagy (115).

8. Ca2+ and autophagy

Cellular response to radiation-induced stress is generally mediated through the production of the second messenger, in-flux of Ca2+ (90,116,117). These radiation-induced changes in Ca2+ concentration can be elevations, oscillations or single transient changes within minutes to days after exposure to radiation. Radiation regulates intracellular Ca2+ concentration through several different pathways. Firstly, radiation can induce the accumulation of ceramide, which can regulate influx of extracellular Ca2+ at plasma membrane channel level (106). Secondly, radiation can activate PLC, the main enzyme localized at the plasma membrane and producing IP3 and consequently inducing Ca2+ release from ER (90,118). Several studies in mammalian cells and yeast models have demonstrated a potential role for mitochondrial parameters regulated by Ca2+ signals in modulating and/or triggering mitophagy (115,119). Ca2+ release events from the ER, the major intracellular Ca2+ -storage organelle that have an immediate effect on the physiological function of mitochondria and lysosomes (120). Ca2+ released from the ER activates members of the PKC family. In mammals, PKCθ, a member of the PKC family, is a novel factor that mediates Ca2+-dependent induction of autophagy in response to ER stress. Increased Ca2+ concentrations induce PKCθ phosporylation and its localization to cytosolic LC3 puncta (121). In addition, the CAMKK2-PRKAA-mTOR pathway is an important signaling pathway for Ca2+ -induced autophagy, which is also a cascade with established roles in multiple cellular processes including ribosome biogenesis and transcription in addition to cell motility and metabolism (119). In addition elevation of intracellular Ca2+ can activate AMPK, which plays a role in Ca2+ -mediated signal transduction pathways (122,123). An overview of the major intracellular autophagic triggers induced by radiation is shown in Fig. 2.

9. Targeting autophagy for radiotherapy

Various preclinical models revealed that autophagy is activated in irradiated tumor cells and that ATG expression patterns are upregulated following irradiation. Inhibition of autophagy genes such as ATG5 alone or a mixture of BECN1, ATG3, ATG4b, and ATG5 together leads to the strongest, and significant sensitizing effect found in radiation (124). Recent in vitro and in vivo studies in preclinical models suggested that modulation of autophagy can be used as a therapeutic modality to enhance the efficacy of radiation therapy. Currently, multiple clinical trials have been initiated combined with autophagy inhibitors, such as hydroxy chloroquine and chloroquine, which associated with increased sensitivity to radiation (125,126). However, the role of autophagy in the resistance of cancer cells to radiation therapy remains controversial. One logical outcome of this argument is that chloroquine and hydroxychloroquine are unlikely to be appropriate drugs for this purpose, because of the fact that patients with malaria are able to endure treatment with these drugs for years, which suggests that the doses of chloroquine and hydroxychloroquine that are used effectively for malaria treatment may not actually be acting to inhibit autophagy. Another logical outcome is that various studies indicated that the sensitivity of tumor cells to radiation could be enhanced by co-treatment with rapamycin, an autophagy promoter, in various kinds of tumor cells (44,50,127).

10. Conclusion

Radiotherapy is one of the best therapeutic choices for cancer treatment (26). Radiation-induced autophagy in cancer cell lines is related to cell death mechanism and autophagy-inducing agents may act especially as radio-sensitizers or radio-resistance (128). Recent studies show that radiation can damage DNA, plasma membrane and cellular organelle such as mitochondria and ER and activate several radiation responsive pathways. During periods of cellular stress, the convergence of these pathways promotes autophagy (129). To clarify cross talk between autophagy and intracellular radiation response, we focused on the proteins and molecules that link the intracellular radiation response with autophagy. A part of these proteins of radiation responsive pathway were verified to participate in the modulation of radiation-induced autophagy. However, others have not been confirmed in the regulation of autophagy. Deep experimental research is needed to further investigate the understanding of the pathways regulating autophagy induced by radiation, which will likely offer new targets for radiotherapy treatment of certain aggressive forms of tumors. Additionally, it would indicate how to regulate the autophagic process or its related pathways that protect cancer cells from undergoing autophagy and may be beneficial to exploit radio-sensitizers. There may also be a therapeutic role for autophagy in protecting normal tissues from lethal radiation exposures. In addition, a recent study showed that autophagy may yield the opposite returns from tumor immunity perspective, irradiation-induced autophagy exerts crucial activity on tumor clearance by the immune system needing further investigation. Clearly, more work is needed to clarify crosstalk between autophagy and intracellular radiation response.

Abbreviations:

AMP

adenosine monophosphate

AMPK

AMP-activated protein kinase

AMBRA1

activating molecule in beclin1-regulated autophagy protein 1

ARF

alternative reading frame protein

ATF6

activating transcription factor 6

ATM

ataxia telangiectasia mutated

ATP

adenosine triphosphate

BNIP3

BCL2 interacting protein 3

CAMKK2

Ca2+/calmodulin-dependent protein kinase kinase 2

CHOP

C/EBP homologous protein

DAPK

death-associated protein kinase

DDR

DNA damage response

DRAM

damage-regulated autophagy modulator

DSBs

DNA double-strand breaks

FIP200

FAK family-interacting protein of 200 kDa

FOXO3a

forkhead box O3a

GTPase

guanosine triphosphatase

HDAC

histone deacetylase

HIF-1α

hypoxia-inducible factor 1α

HMGB1

high-mobility group protein 1

IGF-BP3

insulin-like growth factor-1 binding proteins 3

IRE1

inositol requirement 1

JNK

c-Jun N-terminal kinase

MAPK mitogen activated protein kinase; MDM2

murine double minute 2

mTOR

mechanistic target of rapamycin

LC3

microtubule-associated protein 1 light chain 3

NIX

BCL2/adenovirus E1B 19 kDa interacting protein 3-like

PAS

phagophore assembly site

PERK

protein kinase R-like endoplasmic reticulum kinase

PI3K

phosphatidylinositol 3 kinase

PKC

protein kinase C

PRKAA

protein kinase, AMP activated

PARP-1

poly(ADP-ribose) polymerase-1

PINK1

PTEN induced putative kinase 1

PKC

protein kinase C

PTEN

phosphatase and tensin homolog

PtdIns3K

the class 3 phosphatidylinositol 3-kinase

PI3K

class 1 phosphoinositide 3-kinase

PtdIns3P

phosphatidylinositol 3-phosphate

PUMA

p53-upregulated-modulator-of-apoptosis

RHEB

Ras homolog enriched in brain gene

RNS

reactive nitrogen species

ROS

reactive oxygen species

TSC2

tuberous sclerosis 2

SIRT1

sirtuin1

ULK1

unc-51 like autophagy activating kinase 1

UVRAG

UV radiation resistance-associated gene

VMP1

vacuole membrane protein 1

References

1 

Jaboin JJ, Shinohara ET, Moretti L, Yang ES, Kaminski JM and Lu B: The role of mTOR inhibition in augmenting radiation induced autophagy. Technol Cancer Res Treat. 6:443–447. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Periyasamy P, Guo ML and Buch S: Cocaine induces astrocytosis through ER stress-mediated activation of autophagy. Autophagy. 12:1310–1329. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Codogno P: Shining light on autophagy. Nat Rev Mol Cell Biol. 15:1532014. View Article : Google Scholar : PubMed/NCBI

4 

Park JM, Jung CH, Seo M, Otto NM, Grunwald D, Kim KH, Moriarity B, Kim YM, Starker C, Nho RS, et al: The ULK1 complex mediates MTORC1 signaling to the autophagy initiation machinery via binding and phosphorylating ATG14. Autophagy. 12:547–564. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Yao Z, Delorme-Axford E, Backues SK and Klionsky DJ: Atg41/Icy2 regulates autophagosome formation. Autophagy. 11:2288–2299. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Feng Y, Backues SK, Baba M, Heo JM, Harper JW and Klionsky DJ: Phosphorylation of Atg9 regulates movement to the phagophore assembly site and the rate of autophagosome formation. Autophagy. 12:648–658. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Molejon MI, Ropolo A and Vaccaro MI: VMP1 is a new player in the regulation of the autophagy-specific phosphatidylinositol 3-kinase complex activation. Autophagy. 9:933–935. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Murrow L, Malhotra R and Debnath J: ATG12-ATG3 interacts with Alix to promote basal autophagic flux and late endosome function. Nat Cell Biol. 17:300–310. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Perluigi M, Di Domenico F and Butterfield DA: mTOR signaling in aging and neurodegeneration: At the crossroad between metabolism dysfunction and impairment of autophagy. Neurobiol Dis. 84:39–49. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Yang Z and Klionsky DJ: Mammalian autophagy: Core molecular machinery and signaling regulation. Curr Opin Cell Biol. 22:124–131. 2010. View Article : Google Scholar :

11 

Kroemer G, Mariño G and Levine B: Autophagy and the integrated stress response. Mol Cell. 40:280–293. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Dalby KN, Tekedereli I, Lopez-Berestein G and Ozpolat B: Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy. 6:322–329. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Ito H, Daido S, Kanzawa T, Kondo S and Kondo Y: Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol. 26:1401–1410. 2005.PubMed/NCBI

14 

Li X, Cen Y, Cai Y, Liu T, Liu H, Cao G, Liu D, Li B, Peng W, Zou J, et al: TLR9-ERK-mTOR signaling is critical for autophagic cell death induced by CpG oligodeoxynucleotide 107 combined with irradiation in glioma cells. Sci Rep. 6:271042016. View Article : Google Scholar : PubMed/NCBI

15 

Ko A, Kanehisa A, Martins I, Senovilla L, Chargari C, Dugue D, Mariño G, Kepp O, Michaud M, Perfettini JL, et al: Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ. 21:92–99. 2014. View Article : Google Scholar

16 

Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, et al: Consensus guidelines for the detection of immunogenic cell death. OncoImmunology. 3:e9556912014. View Article : Google Scholar

17 

Mo N, Lu YK, Xie WM, Liu Y, Zhou WX, Wang HX, Nong L, Jia YX, Tan AH, Chen Y, et al: Inhibition of autophagy enhances the radiosensitivity of nasopharyngeal carcinoma by reducing Rad51 expression. Oncol Rep. 32:1905–1912. 2014.PubMed/NCBI

18 

Sun Q, Liu T, Yuan Y, Guo Z, Xie G, Du S, Lin X, Xu Z, Liu M, Wang W, et al: MiR-200c inhibits autophagy and enhances radiosensitivity in breast cancer cells by targeting UBQLN1. Int J Cancer. 136:1003–1012. 2015. View Article : Google Scholar

19 

Yang Y, Yang Y, Yang X, Zhu H, Guo Q, Chen X, Zhang H, Cheng H and Sun X: Autophagy and its function in radiosensitivity. Tumour Biol. 36:4079–4087. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Wu SY, Liu YW, Wang YK, Lin TH, Li YZ, Chen SH and Lee YR: Ionizing radiation induces autophagy in human oral squamous cell carcinoma. J BUON. 19:137–144. 2014.PubMed/NCBI

21 

Saglar E, Unlu S, Babalioglu I, Gokce SC and Mergen H: Assessment of ER stress and autophagy induced by ionizing radiation in both radiotherapy patients and ex vivo irradiated samples. J Biochem Mol Toxicol. 28:413–417. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Bristol ML, Di X, Beckman MJ, Wilson EN, Henderson SC, Maiti A, Fan Z and Gewirtz DA: Dual functions of autophagy in the response of breast tumor cells to radiation: Cytoprotective autophagy with radiation alone and cytotoxic autophagy in radio-sensitization by vitamin D 3. Autophagy. 8:739–753. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Sharma K, Goehe RW, Di X, Hicks MA II, Torti SV, Torti FM, Harada H and Gewirtz DA: A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB 1089. Autophagy. 10:2346–2361. 2014. View Article : Google Scholar

24 

Gewirtz DA, Hilliker ML and Wilson EN: Promotion of autophagy as a mechanism for radiation sensitization of breast tumor cells. Radiother Oncol. 92:323–328. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Sharma K, Le N, Alotaibi M and Gewirtz DA: Cytotoxic autophagy in cancer therapy. Int J Mol Sci. 15:10034–10051. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Huang YH, Yang PM, Chuah QY, Lee YJ, Hsieh YF, Peng CW and Chiu SJ: Autophagy promotes radiation-induced senescence but inhibits bystander effects in human breast cancer cells. Autophagy. 10:1212–1228. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Golden EB, Pellicciotta I, Demaria S, Barcellos-Hoff MH and Formenti SC: The convergence of radiation and immunogenic cell death signaling pathways. Front Oncol. 2:882012. View Article : Google Scholar : PubMed/NCBI

28 

Saitoh T and Akira S: Regulation of innate immune responses by autophagy-related proteins. J Cell Biol. 189:925–935. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Wang Y, Martins I, Ma Y, Kepp O, Galluzzi L and Kroemer G: Autophagy-dependent ATP release from dying cells via lysosomal exocytosis. Autophagy. 9:1624–1625. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G, et al: Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 334:1573–1577. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Ratikan JA, Sayre JW and Schaue D: Chloroquine engages the immune system to eradicate irradiated breast tumors in mice. Int J Radiat Oncol Biol Phys. 87:761–768. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Havaki S, Kotsinas A, Chronopoulos E, Kletsas D, Georgakilas A and Gorgoulis VG: The role of oxidative DNA damage in radiation induced bystander effect. Cancer Lett. 356:43–51. 2015. View Article : Google Scholar

33 

Sengupta S and Harris CC: p53: traffic cop at the crossroads of DNA repair and recombination. Nat Rev Mol Cell Biol. 6:44–55. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Smith J, Tho LM, Xu N and Gillespie DA: The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res. 108:73–112. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Liang N, Jia L, Liu Y, Liang B, Kong D, Yan M, Ma S and Liu X: ATM pathway is essential for ionizing radiation-induced autophagy. Cell Signal. 25:2530–2539. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Jen KY and Cheung VG: Identification of novel p53 target genes in ionizing radiation response. Cancer Res. 65:7666–7673. 2005.PubMed/NCBI

37 

Li M, Brooks CL, Wu-Baer F, Chen D, Baer R and Gu W: Mono-versus polyubiquitination: Differential control of p53 fate by Mdm2. Science. 302:1972–1975. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Fei P and El-Deiry WS: P53 and radiation responses. Oncogene. 22:5774–5783. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Tang J, Di J, Cao H, Bai J and Zheng J: p53-mediated autophagic regulation: A prospective strategy for cancer therapy. Cancer Lett. 363:101–107. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Zheng R, Yao Q, Du S, Ren C, Sun Q, Xu Z, Lin X and Yuan Y: The status of p53 in cancer cells affects the role of autophagy in tumor radiosensitisation. J BUON. 19:336–341. 2014.PubMed/NCBI

41 

Feng Z, Zhang H, Levine AJ and Jin S: The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci USA. 102:8204–8209. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Cui L, Song Z, Liang B, Jia L, Ma S and Liu X: Radiation induces autophagic cell death via the p53/DRAM signaling pathway in breast cancer cells. Oncol Rep. 35:3639–3647. 2016.PubMed/NCBI

43 

Xu K, Liu P and Wei W: mTOR signaling in tumorigenesis. Biochim Biophys Acta. 1846:638–654. 2014.PubMed/NCBI

44 

Zheng H, Wang M, Wu J, Wang ZM, Nan HJ and Sun H: Inhibition of mTOR enhances radiosensitivity of lung cancer cells and protects normal lung cells against radiation. Biochem Cell Biol. 94:213–220. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, et al: Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 331:456–461. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Kim J, Kundu M, Viollet B and Guan KL: AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 13:132–141. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Hönscheid P, Datta K and Muders MH: Autophagy: Detection, regulation and its role in cancer and therapy response. Int J Radiat Biol. 90:628–635. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Nagata Y, Takahashi A, Ohnishi K, Ota I, Ohnishi T, Tojo T and Taniguchi S: Effect of rapamycin, an mTOR inhibitor, on radiation sensitivity of lung cancer cells having different p53 gene status. Int J Oncol. 37:1001–1010. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Dai ZJ, Gao J, Kang HF, Ma YG, Ma XB, Lu WF, Lin S, Ma HB, Wang XJ and Wu WY: Targeted inhibition of mammalian target of rapamycin (mTOR) enhances radiosensitivity in pancreatic carcinoma cells. Drug Des Devel Ther. 7:149–159. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Zhang D, Xiang J, Gu Y, Xu W, Xu H, Zu M, Pei D and Zheng J: Inhibition of mammalian target of rapamycin by rapamycin increases the radiosensitivity of esophageal carcinoma Eca109 cells. Oncol Lett. 8:575–581. 2014.PubMed/NCBI

51 

Ushijima H, Suzuki Y, Oike T, Komachi M, Yoshimoto Y, Ando K, Okonogi N, Sato H, Noda SE, Saito J, et al: Radiosensitization effect of an mTOR inhibitor, temsirolimus, on lung adenocarcinoma A549 cells under normoxic and hypoxic conditions. J Radiat Res (Tokyo). 56:663–668. 2015. View Article : Google Scholar

52 

Tsai WB, Chung YM, Takahashi Y, Xu Z and Hu MC: Functional interaction between FOXO3a and ATM regulates DNA damage response. Nat Cell Biol. 10:460–467. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Yang JY, Xia W and Hu MC: Ionizing radiation activates expression of FOXO3a, Fas ligand, and Bim, and induces cell apoptosis. Int J Oncol. 29:643–648. 2006.PubMed/NCBI

54 

Tarrade S, Bhardwaj T, Flegal M, Bertrand L, Velegzhaninov I, Moskalev A and Klokov D: Histone H2AX is involved in FoxO3a-mediated transcriptional responses to ionizing radiation to maintain genome stability. Int J Mol Sci. 16:29996–30014. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Tran H, Brunet A, Grenier JM, Datta SR, Fornace AJ Jr, DiStefano PS, Chiang LW and Greenberg ME: DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science. 296:530–534. 2002. View Article : Google Scholar : PubMed/NCBI

56 

Cho EA, Kim EJ, Kwak SJ and Juhnn YS: cAMP signaling inhibits radiation-induced ATM phosphorylation leading to the augmentation of apoptosis in human lung cancer cells. Mol Cancer. 13:362014. View Article : Google Scholar : PubMed/NCBI

57 

Rodríguez-Vargas JM, Ruiz-Magaña MJ, Ruiz-Ruiz C, Majuelos-Melguizo J, Peralta-Leal A, Rodríguez MI, Muñoz-Gámez JA, de Almodóvar MR, Siles E, Rivas AL, et al: ROS-induced DNA damage and PARP-1 are required for optimal induction of starvation-induced autophagy. Cell Res. 22:1181–1198. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Yang M, Liu L, Xie M, Sun X, Yu Y, Kang R, Yang L, Zhu S, Cao L and Tang D: Poly-ADP-ribosylation of HMGB1 regulates TNFSF10/TRAIL resistance through autophagy. Autophagy. 11:214–224. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Bridges KA, Toniatti C, Buser CA, Liu H, Buchholz TA and Meyn RE: Niraparib (MK-4827), a novel poly(ADP-Ribose) polymerase inhibitor, radiosensitizes human lung and breast cancer cells. Oncotarget. 5:5076–5086. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Chen ZT, Zhao W, Qu S, Li L, Lu XD, Su F, Liang ZG, Guo SY and Zhu XD: PARP-1 promotes autophagy via the AMPK/mTOR pathway in CNE-2 human nasopharyngeal carcinoma cells following ionizing radiation, while inhibition of autophagy contributes to the radiation sensitization of CNE-2 cells. Mol Med Rep. 12:1868–1876. 2015.PubMed/NCBI

61 

Xie Y, Zhang J, Ye S, He M, Ren R, Yuan D and Shao C: SirT1 regulates radiosensitivity of hepatoma cells differently under normoxic and hypoxic conditions. Cancer Sci. 103:1238–1244. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Lapierre LR, Kumsta C, Sandri M, Ballabio A and Hansen M: Transcriptional and epigenetic regulation of autophagy in aging. Autophagy. 11:867–880. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Zhang B, Davidson MM, Zhou H, Wang C, Walker WF and Hei TK: Cytoplasmic irradiation results in mitochondrial dysfunction and DRP1-dependent mitochondrial fission. Cancer Res. 73:6700–6710. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Kam WW and Banati RB: Effects of ionizing radiation on mitochondria. Free Radic Biol Med. 65:607–619. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Shadel GS and Horvath TL: Mitochondrial ROS signaling in organismal homeostasis. Cell. 163:560–569. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Lin WJ and Kuang HY: Oxidative stress induces autophagy in response to multiple noxious stimuli in retinal ganglion cells. Autophagy. 10:1692–1701. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Filomeni G, De Zio D and Cecconi F: Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 22:377–388. 2015. View Article : Google Scholar :

68 

Zhang D, Wang W, Sun X, Xu D, Wang C, Zhang Q, Wang H, Luo W, Chen Y, Chen H, et al: AMPK regulates autophagy by phosphorylating BECN1 at Threonine 388. Autophagy. 12:1447–1459. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Shimura T, Kobayashi J, Komatsu K and Kunugita N: Severe mitochondrial damage associated with low-dose radiation sensitivity in ATM- and NBS1-deficient cells. Cell Cycle. 15:1099–1107. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Shimura T, Sasatani M, Kamiya K, Kawai H, Inaba Y and Kunugita N: Mitochondrial reactive oxygen species perturb AKT/cyclin D1 cell cycle signaling via oxidative inactivation of PP2A in lowdose irradiated human fibroblasts. Oncotarget. 7:3559–3570. 2016.

71 

Garg AD, Dudek AM, Ferreira GB, Verfaillie T, Vandenabeele P, Krysko DV, Mathieu C and Agostinis P: ROS-induced autophagy in cancer cells assists in evasion from determinants of immunogenic cell death. Autophagy. 9:1292–1307. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Clerkin JS, Naughton R, Quiney C and Cotter TG: Mechanisms of ROS modulated cell survival during carcinogenesis. Cancer Lett. 266:30–36. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Datta K, Suman S and Fornace AJ Jr: Radiation persistently promoted oxidative stress, activated mTOR via PI3K/Akt, and downregulated autophagy pathway in mouse intestine. Int J Biochem Cell Biol. 57:167–176. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR and Youle RJ: PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 8:e10002982010. View Article : Google Scholar : PubMed/NCBI

75 

Poillet-Perez L, Despouy G, Delage-Mourroux R and Boyer-Guittaut M: Interplay between ROS and autophagy in cancer cells, from tumor initiation to cancer therapy. Redox Biol. 4:184–192. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Zhang B, Wang Y, Pang X, Su Y, Ai G and Wang T: ER stress induced by ionising radiation in IEC-6 cells. Int J Radiat Biol. 86:429–435. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Li F, Zheng X, Liu Y, Li P, Liu X, Ye F, Zhao T, Wu Q, Jin X and Li Q: Different roles of CHOP and JNK in mediating radiation-induced autophagy and apoptosis in breast cancer cells. Radiat Res. 185:539–548. 2016. View Article : Google Scholar : PubMed/NCBI

78 

Chiu HW, Fang WH, Chen YL, Wu MD, Yuan GF, Ho SY and Wang YJ: Monascuspiloin enhances the radiation sensitivity of human prostate cancer cells by stimulating endoplasmic reticulum stress and inducing autophagy. PLoS One. 7:e404622012. View Article : Google Scholar : PubMed/NCBI

79 

Chiu HW, Yeh YL, Wang YC, Huang WJ, Ho SY, Lin P and Wang YJ: Combination of the novel histone deacetylase inhibitor YCW1 and radiation induces autophagic cell death through the downregulation of BNIP3 in triple-negative breast cancer cells in vitro and in an orthotopic mouse model. Mol Cancer. 15:462016. View Article : Google Scholar : PubMed/NCBI

80 

Yang Z, Xu Y, Xu L, Maccauro G, Rossi B, Chen Y, Li H, Zhang J, Sun H, Yang Y, et al: Regulation of autophagy via PERK-eIF2α effectively relieve the radiation myelitis induced by iodine-125. PLoS One. 8:e768192013. View Article : Google Scholar

81 

Kim KW, Moretti L, Mitchell LR, Jung DK and Lu B: Endoplasmic reticulum stress mediates radiation-induced autophagy by perk-eIF2alpha in caspase-3/7-deficient cells. Oncogene. 29:3241–3251. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Kim EJ, Lee YJ, Kang S and Lim YB: Ionizing radiation activates PERK/eIF2α/ATF4 signaling via ER stress-independent pathway in human vascular endothelial cells. Int J Radiat Biol. 90:306–312. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, Yun C, Popko B, Paules R, et al: An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 11:619–633. 2003. View Article : Google Scholar : PubMed/NCBI

84 

Milani M, Rzymski T, Mellor HR, Pike L, Bottini A, Generali D and Harris AL: The role of ATF4 stabilization and autophagy in resistance of breast cancer cells treated with Bortezomib. Cancer Res. 69:4415–4423. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Huang Q, Wu YT, Tan HL, Ong CN and Shen HM: A novel function of poly(ADP-ribose) polymerase-1 in modulation of autophagy and necrosis under oxidative stress. Cell Death Differ. 16:264–277. 2009. View Article : Google Scholar

86 

Kyriakis JM, Banerjee P, Nikolakaki E, Dai T, Rubie EA, Ahmad MF, Avruch J and Woodgett JR: The stress-activated protein kinase subfamily of c-Jun kinases. Nature. 369:156–160. 1994. View Article : Google Scholar : PubMed/NCBI

87 

Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP and Ron D: Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 287:664–666. 2000. View Article : Google Scholar : PubMed/NCBI

88 

Senft D and Ronai ZA: UPR, autophagy, and mitochondria crosstalk underlies the ER stress response. Trends Biochem Sci. 40:141–148. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Davalli P, Mitic T, Caporali A, Lauriola A and D’Arca D: ROS, cell senescence, and novel molecular mechanisms in aging and age-related diseases. Oxid Med Cell Longev. 2016:35651272016. View Article : Google Scholar : PubMed/NCBI

90 

Corre I, Niaudet C and Paris F: Plasma membrane signaling induced by ionizing radiation. Mutat Res. 704:61–67. 2010. View Article : Google Scholar : PubMed/NCBI

91 

Liu Y, Cui Y, Shi M, Zhang Q, Wang Q and Chen X: Deferoxamine promotes MDA-MB-231 cell migration and invasion through increased ROS-dependent HIF-1α accumulation. Cell Physiol Biochem. 33:1036–1046. 2014. View Article : Google Scholar

92 

Sridharan S, Jain K and Basu A: Regulation of autophagy by kinases. Cancers (Basel). 3:2630–2654. 2011. View Article : Google Scholar

93 

Bode JG, Ehlting C and Häussinger D: The macrophage response towards LPS and its control through the p38(MAPK)-STAT3 axis. Cell Signal. 24:1185–1194. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Tang G, Yue Z, Talloczy Z, Hagemann T, Cho W, Messing A, Sulzer DL and Goldman JE: Autophagy induced by Alexander disease-mutant GFAP accumulation is regulated by p38/MAPK and mTOR signaling pathways. Hum Mol Genet. 17:1540–1555. 2008. View Article : Google Scholar : PubMed/NCBI

95 

Lien SC, Chang SF, Lee PL, Wei SY, Chang MD, Chang JY and Chiu JJ: Mechanical regulation of cancer cell apoptosis and autophagy: Roles of bone morphogenetic protein receptor, Smad1/5, and p38 MAPK. Biochim Biophys Acta. 1833:3124–3133. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Kim YH, Yoo KC, Cui YH, Uddin N, Lim EJ, Kim MJ, Nam SY, Kim IG, Suh Y and Lee SJ: Radiation promotes malignant progression of glioma cells through HIF-1alpha stabilization. Cancer Lett. 354:132–141. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Gu Q, He Y, Ji J, Yao Y, Shen W, Luo J, Zhu W, Cao H, Geng Y, Xu J, et al: Hypoxia-inducible factor 1α (HIF-1α) and reactive oxygen species (ROS) mediates radiation-induced invasiveness through the SDF-1α/CXCR4 pathway in non-small cell lung carcinoma cells. Oncotarget. 6:10893–10907. 2015. View Article : Google Scholar : PubMed/NCBI

98 

Li P, Shi J, He Q, Hu Q, Wang YY, Zhang LJ, Chan WT and Chen WX: Streptococcus pneumoniae induces autophagy through the inhibition of the PI3K-I/Akt/mTOR pathway and ROS hypergeneration in A549 cells. PLoS One. 10:e01227532015. View Article : Google Scholar : PubMed/NCBI

99 

Wang GL, Jiang BH, Rue EA and Semenza GL: Hypoxiainducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA. 92:5510–5514. 1995. View Article : Google Scholar

100 

Harada H: Hypoxia-inducible factor 1-mediated characteristic features of cancer cells for tumor radioresistance. J Radiat Res (Tokyo). 57(Suppl 1): i99–i105. 2016. View Article : Google Scholar

101 

Koshikawa N, Hayashi J, Nakagawara A and Takenaga K: Reactive oxygen species-generating mitochondrial DNA mutation up-regulates hypoxia-inducible factor-1alpha gene transcription via phosphatidylinositol 3-kinase-Akt/protein kinase C/histone deacetylase pathway. J Biol Chem. 284:33185–33194. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Bonello S, Zähringer C, BelAiba RS, Djordjevic T, Hess J, Michiels C, Kietzmann T and Görlach A: Reactive oxygen species activate the HIF-1alpha promoter via a functional NFkappaB site. Arterioscler Thromb Vasc Biol. 27:755–761. 2007. View Article : Google Scholar : PubMed/NCBI

103 

Noman MZ, Janji B, Berchem G, Mami-Chouaib F and Chouaib S: Hypoxia-induced autophagy: A new player in cancer immunotherapy? Autophagy. 8:704–706. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, Pouysségur J and Mazure NM: Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol. 29:2570–2581. 2009. View Article : Google Scholar : PubMed/NCBI

105 

Tirodkar TS and Voelkel-Johnson C: Sphingolipids in apoptosis. Exp Oncol. 34:231–242. 2012.PubMed/NCBI

106 

Aureli M, Murdica V, Loberto N, Samarani M, Prinetti A, Bassi R and Sonnino S: Exploring the link between ceramide and ionizing radiation. Glycoconj J. 31:449–459. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Young MM, Kester M and Wang HG: Sphingolipids: Regulators of crosstalk between apoptosis and autophagy. J Lipid Res. 54:5–19. 2013. View Article : Google Scholar :

108 

Edinger AL: Starvation in the midst of plenty: Making sense of ceramide-induced autophagy by analysing nutrient transporter expression. Biochem Soc Trans. 37:253–258. 2009. View Article : Google Scholar : PubMed/NCBI

109 

Peralta ER and Edinger AL: Ceramide-induced starvation triggers homeostatic autophagy. Autophagy. 5:407–409. 2009. View Article : Google Scholar : PubMed/NCBI

110 

Li DD, Wang LL, Deng R, Tang J, Shen Y, Guo JF, Wang Y, Xia LP, Feng GK, Liu QQ, et al: The pivotal role of c-Jun NH2-terminal kinase-mediated Beclin 1 expression during anticancer agents-induced autophagy in cancer cells. Oncogene. 28:886–898. 2009. View Article : Google Scholar

111 

Jiang W and Ogretmen B: Autophagy paradox and ceramide. Biochim Biophys Acta. 1841:783–792. 2014. View Article : Google Scholar :

112 

Dany M and Ogretmen B: Ceramide induced mitophagy and tumor suppression. Biochim Biophys Acta. 1853B:2834–2845. 2015. View Article : Google Scholar

113 

Salazar M, Carracedo A, Salanueva IJ, Hernández-Tiedra S, Lorente M, Egia A, Vázquez P, Blázquez C, Torres S, García S, et al: Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest. 119:1359–1372. 2009. View Article : Google Scholar : PubMed/NCBI

114 

Sentelle RD, Senkal CE, Jiang W, Ponnusamy S, Gencer S, Selvam SP, Ramshesh VK, Peterson YK, Lemasters JJ, Szulc ZM, et al: Ceramide targets autophagosomes to mitochondria and induces lethal mitophagy. Nat Chem Biol. 8:831–838. 2012. View Article : Google Scholar : PubMed/NCBI

115 

Rimessi A, Bonora M, Marchi S, Patergnani S, Marobbio CM, Lasorsa FM and Pinton P: Perturbed mitochondrial Ca2+ signals as causes or consequences of mitophagy induction. Autophagy. 9:1677–1686. 2013. View Article : Google Scholar : PubMed/NCBI

116 

Voehringer DW, Story MD, O’Neil RG and Meyn RE: Modulating Ca2+ in radiation-induced apoptosis suppresses DNA fragmentation but does not enhance clonogenic survival. Int J Radiat Biol. 71:237–243. 1997. View Article : Google Scholar : PubMed/NCBI

117 

Teshima K, Yamamoto A, Yamaoka K, Honda Y, Honda S, Sasaki T and Kojima S: Involvement of calcium ion in elevation of mRNA for gamma-glutamylcysteine synthetase (gamma-GCS) induced by low-dose gamma-rays. Int J Radiat Biol. 76:1631–1639. 2000. View Article : Google Scholar

118 

Todd DG, Mikkelsen RB, Rorrer WK, Valerie K and Schmidt-Ullrich RK: Ionizing radiation stimulates existing signal transduction pathways involving the activation of epidermal growth factor receptor and ERBB-3, and changes of intracellular calcium in A431 human squamous carcinoma cells. J Recept Signal Transduct Res. 19:885–908. 1999. View Article : Google Scholar : PubMed/NCBI

119 

East DA and Campanella M: Ca2+ in quality control: An unresolved riddle critical to autophagy and mitophagy. Autophagy. 9:1710–1719. 2013. View Article : Google Scholar : PubMed/NCBI

120 

La Rovere RM, Roest G, Bultynck G and Parys JB: Intracellular Ca(2+) signaling and Ca(2+) microdomains in the control of cell survival, apoptosis and autophagy. Cell Calcium. 60:74–87. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Sakaki K, Wu J and Kaufman RJ: Protein kinase Ctheta is required for autophagy in response to stress in the endoplasmic reticulum. J Biol Chem. 283:15370–15380. 2008. View Article : Google Scholar : PubMed/NCBI

122 

Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, Carlson M and Carling D: Ca2+ /calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2:21–33. 2005. View Article : Google Scholar : PubMed/NCBI

123 

Zhang J, Chiu J, Zhang H, Qi T, Tang Q, Ma K, Lu H and Li G: Autophagic cell death induced by resveratrol depends on the Ca(2+)/AMPK/mTOR pathway in A549 cells. Biochem Pharmacol. 86:317–328. 2013. View Article : Google Scholar : PubMed/NCBI

124 

Apel A, Herr I, Schwarz H, Rodemann HP and Mayer A: Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 68:1485–1494. 2008. View Article : Google Scholar : PubMed/NCBI

125 

Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S, Mikkelson T, Wang D, Chang YC, Hu J, et al: A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy. 10:1359–1368. 2014. View Article : Google Scholar : PubMed/NCBI

126 

Rojas-Puentes LL, Gonzalez-Pinedo M, Crismatt A, Ortega-Gomez A, Gamboa-Vignolle C, Nuñez-Gomez R, Dorantes-Gallareta Y, Arce-Salinas C and Arrieta O: Phase II randomized, double-blind, placebo-controlled study of wholebrain irradiation with concomitant chloroquine for brain metastases. Radiat Oncol. 8:2092013. View Article : Google Scholar

127 

Chen YH, Wei MF, Wang CW, Lee HW, Pan SL, Gao M, Kuo SH, Cheng AL and Teng CM: Dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor is an effective radiosensitizer for colorectal cancer. Cancer Lett. 357:582–590. 2015. View Article : Google Scholar

128 

Ozpolat B and Benbrook DM: Targeting autophagy in cancer management - strategies and developments. Cancer Manag Res. 7:291–299. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Liang DH, El-Zein R and Dave B: Autophagy inhibition to increase radiosensitization in breast cancer. J Nucl Med Radiat Ther. 6:62015. View Article : Google Scholar

Related Articles

Journal Cover

December-2016
Volume 49 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hu L, Wang H, Huang L, Zhao Y and Wang J: Crosstalk between autophagy and intracellular radiation response (Review). Int J Oncol 49: 2217-2226, 2016
APA
Hu, L., Wang, H., Huang, L., Zhao, Y., & Wang, J. (2016). Crosstalk between autophagy and intracellular radiation response (Review). International Journal of Oncology, 49, 2217-2226. https://doi.org/10.3892/ijo.2016.3719
MLA
Hu, L., Wang, H., Huang, L., Zhao, Y., Wang, J."Crosstalk between autophagy and intracellular radiation response (Review)". International Journal of Oncology 49.6 (2016): 2217-2226.
Chicago
Hu, L., Wang, H., Huang, L., Zhao, Y., Wang, J."Crosstalk between autophagy and intracellular radiation response (Review)". International Journal of Oncology 49, no. 6 (2016): 2217-2226. https://doi.org/10.3892/ijo.2016.3719