Next Article in Journal
Protein Oxidative Damage in UV-Related Skin Cancer and Dysplastic Lesions Contributes to Neoplastic Promotion and Progression
Next Article in Special Issue
Autophagy: New Insights into Its Roles in Cancer Progression and Drug Resistance
Previous Article in Journal
The Application of Arsenic Trioxide in Ameliorating ABT-737 Target Therapy on Uterine Cervical Cancer Cells through Unique Pathways in Cell Death
Previous Article in Special Issue
Molecular Mechanisms Underlying Autophagy-Mediated Treatment Resistance in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Determinants of Cancer Therapy Resistance to HDAC Inhibitor-Induced Autophagy

by
Maria Mrakovcic
1,2,* and
Leopold F. Fröhlich
1
1
Department of Cranio-Maxillofacial Surgery, University of Münster, 48149 Münster, Germany
2
Department of Medical Microbiology, University of Münster, 48149 Münster, Germany
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(1), 109; https://doi.org/10.3390/cancers12010109
Submission received: 22 November 2019 / Revised: 16 December 2019 / Accepted: 20 December 2019 / Published: 31 December 2019
(This article belongs to the Special Issue The Role of Autophagy in Cancer Progression and Drug Resistance)

Abstract

:
Histone deacetylation inhibitors (HDACi) offer high potential for future cancer therapy as they can re-establish the expression of epigenetically silenced cell death programs. HDACi-induced autophagy offers the possibility to counteract the frequently present apoptosis-resistance as well as stress conditions of cancer cells. Opposed to the function of apoptosis and necrosis however, autophagy activated in cancer cells can engage in a tumor-suppressive or tumor-promoting manner depending on mostly unclarified factors. As a physiological adaption to apoptosis resistance in early phases of tumorigenesis, autophagy seems to resume a tumorsuppressive role that confines tumor necrosis and inflammation or even induces cell death in malignant cells. During later stages of tumor development, chemotherapeutic drug-induced autophagy seems to be reprogrammed by the cancer cell to prevent its elimination and support tumor progression. Consistently, HDACi-mediated activation of autophagy seems to exert a protective function that prevents the induction of apoptotic or necrotic cell death in cancer cells. Thus, resistance to HDACi-induced cell death is often encountered in various types of cancer as well. The current review highlights the different mechanisms of HDACi-elicited autophagy and corresponding possible molecular determinants of therapeutic resistance in cancer.

Graphical Abstract

1. Introduction: Autophagy

Autophagy is an evolutionary well conserved cellular process in eukaryotes that was established already in yeast and has developed to a highly sophisticated version in mammals [1,2,3]. The basic physiologic function of autophagy is to retrieve cellular components, such as denatured, aged, damaged, or non-required molecules and organelles. This mechanism is not only important under normal physiological conditions where it occurs at basal levels for sustaining proper homeostasis (e.g., during development and differentiation) and viability but also predominantly during times of nutrient deprivation and other stressfull settings (e.g., starvation, hypoxia, oxidative stress) [4,5,6,7]. Principally, autophagy is categorized into the classes of macroautophagy, microautophagy, and chaperone-mediated autophagy, depending on the engaged pathway [8,9,10]. Furthermore, autophagy can occur either selective or nonselective, meaning that cargo for sequestration is targeted specifically e.g., via autophagy receptors such as Sqstm1/p62 or randomly e.g., bulk cytoplasm [11]. While discovery of the molecular mechanisms governing of the non-selective starvation-induced mode of autophagy can be attributed to the Nobel prize-awarded research of Yoshinori Ohsumi, it was Daniel J. Klionsky´s group that revealed the molecular process of selective autophagy by defining the cytoplasm-to-vacuole targeting (CVT) pathway, a few years later [1,12,13]. Nevertheless, the term autophagy (from the ancient Greek “aὐsόφacος“, meaning “self-eating”) was initially forged by the Belgian cytologist and biochemist Christian De Duve in 1963 for describing intracellular vesicles that contain parts of the cytoplasm and organelles in various states of disintegration [10,14].
In general, the process of autophagy is a highly regulated process including a set of signaling cascades. Nevertheless, a complex genetic program required in cellular homeostasis or cell death is still not exhaustedly explored and might also differ between basal autophagy and stimulus-induced autophagy [1,6]. General triggering signals of autophagy are very heterogeneous and comprise for instance the shortage of nutrients, the presence of aggregated protein, reactive oxygen species (ROS) in the cell, or damaged cellular organelles [15,16,17]. Aged or damaged cellular components are degraded and retained in specifically for this process allocated autophagosomes. The event of macroautophagy, representing the best investigated form of the cellular degradation program, is therefore most often simply called autophagy and describes the controlled formation of an autophagosome. It represents a vesicle with a double-layered membrane that forms around the targeted cellular component and thereby allows its separation from the remaining cytoplasm [18]. In a transitory stage, subsequent maturation and fusion of this vesicle with lysosomes, exposes the vesicle cargo to lysosomal enzyme degradation by cathepsin and calpain proteases, and redelivers it to the cell for recycling [19]. Vesicles formed in this way are then called autophagolysosomes. Taken together, this dynamic process that includes the formation, engulfment, and lysosomal fusion of autophagosomes is commonly described by the term “autophagic flux” and is defined as a measure of autophagic degradation activity [11,20,21].
Initially, autophagosomes are formed at the phagophore assembly site and was alternatively termed pre-autophagosomal structure (PAS) [22]. Presently, it is concluded that this initial structure, that recruits various autophagy-related (ATG) proteins, is derived from the endoplasmic reticulum through the formation of the omegasome [23,24]. ATG proteins, entail a specific sequence of events in formation, growth, and closure of the autophagosome and have been summarized into five stages [25]. These involve the ATG1/ULK kinase complex during initiation, the ATG12 conjugation system during nucleation, the ATG8/LC3 conjugation/deconjugation system during elongation, the phosphatidylinositol 3-kinase complex during maturation, and the ATG9/ATG9L1 cycling system during degradation.

2. The Oncogenic Role of Autophagy

As autophagy represents a physiological adaptive strategy to stressfull conditions, its disruption was found to contribute to the pathogenesis of disease, such as cancer, by promoting and accelerating tumorigenesis [17,26,27,28]. Although this cellular process has been observed for half a century, the exploration of its significance for cancer development and progression has only recen tly been initiated with the identification of master regulators, such as ATG genes. In this regard, the role of autophagy in cancer holds a high potential for future therapy and is consequently a matter of intensive investigation in laboratory and in clinical studies. However, opposed to other forms of cell death such as apoptosis, autophagy has been characterized by an ambiguous role in cancer in reply to adverse stress events as it supports either an oncosuppressive or an oncosupportive mode of response [29,30,31]. Thus, on the one hand, autophagy may help in the elimination of oncogenic factors, such as oncoproteins, but on the other hand autophagy might be activated as a prosurvival response to cancer treatment, thereby reducing therapeutic efficiency. The most often unpredictable survival- or death-promoting outcome of autophagy thereby seems to rely on the duration and dose of drugs, on the type and stage of cancer, as well as its genetic predisposition [29,32,33,34,35,36,37,38]. The later fact might prevailingly relate to the mutational effects of the most commonly altered tumor suppressor genes (such as LKB-1, PTEN, BECLIN-1, TP53, FOXO1, DAPK1) or oncogenes (such as BCL-2 family genes, PI3K, AKT, mTOR, RAS) that together with microRNAs represent the main regulators of autophagy [39,40]. Of note, microRNAs, belonging to the cellular epigenome, have been evidenced to modify the activity of mRNAs encoding for key autophagy-promoting proteins such as PI3K, ULK1, Beclin-1, LC3B, ATG4 family members, ATG5, ATG7, ATG10, ATG12, or ATG16L1 [41]. Nevertheless, further factors might be found complementing the incomplete mechanistic insights concerning autophagic signaling pathways; these will presumably contribute to the transcriptional and epigenetic regulation of the complex autophagic response, resulting in cell survival or cell death triggered by disease or pharmaceutical intervention. Thus, in addition to the relatively few experimentally validated autophagy-specific transcription factors binding to the promoters of autophagic regulatory proteins, even more transcription factor interacting binding sites are predicted by bioinformatics algorithms [30]. Additionally, selective autophagy influencing the recruitment and degradation of cell survival factors in autophagosomes such as the take-up of catalase from the cytoplasm that would induce ROS generation and induction of cell death might alter the fate of the autophagic response [42,43]. Selective autophagy is a tighly regulated process that depends on autophagy receptors such as Sqstm1/p62 and NBR1 that are controlled by posttranslational modifications and connect the mostly ubiquitin-labeled cargo to proteins of the ATG8 family. These act as adaptors that attach cargo to the inner surface of the growing phagophore.
The suppressive or supportive mode of autophagy was tightlyconnected to a function of time and differentiation during tumor development. Thus, during initial phases of tumorigenesis the protective function of autophagy prevails by removing damaging agents from the cell thereby decreasing the tendency of damaged cells to transform into tumor cells. As an example, autophagy was documented to prevent the increased effects of oxidative stress by clearing damaged organelles at the cellular level [44]. Supportive models for this tumor-suppressive action of autophagy are found in hemizygous Beclin-1-deficient mice that lose their autophagic regulatory potential thereby being increasingly susceptible to tumor formation [28,45]. During later phases of tumorigenesis however, autophagy seems to be reprogrammed by the tumor cell to prevent its eradication and even support tumor progression and metastasis. For example, autophagy can help to reduce ROS-induced radical formation effects of metabolic stress products that would harm the tumor cell and supply it with nutrients thereby enhancing tumor survival [46]. These pathological circumstances however offer the possibility to expedite and overstress the autophagic program by pharmacological interference via unknown mechanisms and direct the tumor-promoting conditions towards induction of cell death. Prolonged autophagy thereby seems to deplete critical survival factors or eliminate essential cellular contents and organelles which could also promote cell death by activating apoptosis or necroptosis. Particularly, as a physiological adaption to apoptosis resistance in tumor cells, autophagy resumes a tumorsuppressive role, that confines inflammation and tumor necrosis [47,48]. This could be verified in our own model of the apoptosis-resistant uterine sarcoma cell line ESS-1; in contrast to suberoylanilide hydroxamic acid (SAHA)-inducible apoptotic cell death in the cell line MES-SA, ESS-1 was found to undergo autophagy-associated cell death due to a homozygous nonsense mutation in the TP53 gene that causes p53 protein deficiency or degradation [49,50].
Autophagy is furthermore activated in response to an increasing number of drugs used in cancer treatment to protect against cellular stress. This protective function of autophagy can be considered as a mutual response of the cell that prevents both, induction of either apoptotic or necrotic cell death [36,51,52]. As a quite often realized problem, however, autophagy also facilitates resistance of the tumor cell to chemotherapy and radiation treatment [46,53]. To avoid and re-sensitize therapeutic resistant cancer cells, several strategies deploy the use of inhibitors as an adjuvant therapy, such as 3-methyladenine (3-MA), the lysosomotropic substance chloroquine (CQ) or its analog, hydroxychloroquine (HCQ), as well as BafA1 (bafilomycin A1), or use gene silencing that abrogate autophagy [54,55,56,57]. In fact, drug mediated inhibition of autophagy in combination with cytotoxic drugs constitutes a novel potential target in cancer therapy [31,47,58,59,60,61,62]. Nevertheless, in the case of histone deacetylase inhibitor (HDACi) treatment which is intended to promote alternative modes of cell death this can result in a drawback; tevidently, in the presence of apoptosis resistance, disruption of autophagy, will evoke enhanced tumor survival as the only escape route is blocked. This should again underline the necessity of affirming the context-dependent role of autophagy in cancer treatment by previous investigation of the limiting molecular determinants, such as oncosuppressors and oncoproteins [37,47,63,64,65].

3. Autophagy as a Therapeutic Target in Drug Resistance

Increasing evidence implicates an important role of autophagy in developing resistance to chemotherapeutic treatment of cancers [37,66,67,68,69,70]. This is a particular drawback in the therapy of tumors that are anyway hard to treat. A multiple spectrum of different cancer drugs has been demonstrated to activate autophagy which in several cases by eliciting therapeutic resistance also facilitates survival of tumor cells [31,63]. Thus, interrelated autophagy induction and drug resistance have been observed in ovarian cancer treated with paclitaxel, in ovarian and esophageal cancer teated with cisplatin, in lung cancer following hypoxia, in melanoma treated with cyclin dependent kinase (CDK) inhibitors, or in glioblatoma cells treated with the HDACiTSA, to just name a few (reviewed in [31]).
Acquired resistance will be indicated by increased levels of autophagy that was indeed observed in several cases, and therefore can be tackled by the additional use of autophagy inhibitors such as CQ. In addition to pharmacological inhibitors, re-sensitization of autophagy can also be regained by genetic inhibitors, which could target several different concerned pathways of acquired autophagic resistance as documented in vitro and by pre-clinical studies [63]. This strategy can either target specific pathways of autophagy or block the entire process to obtain a clinical benefit. Specifically, due to the frequent link between drug-mediated induction of autophagy and chemotherapeutic resistance, the suppression of autophagy presents in return most evidently a promising target in cancer therapy. For example, in estrogen receptor-positive breast cancer, inhibition of autophagy sensitized resistant tumors to tamoxifen-induced killing [71,72]. Similarly, in prostate cancer, autophagy inhibition overcame resistance to enzalutamide [73]. Autophagy is also induced in response to treatment of gastrointestinal stromal tumor (GIST) cells with Imatinib™ and inhibition of autophagy (by CQ) caused tumor cell apoptosis [74].
As one of the most obvious molecular explanations for the chemotherapy-associated activation of autophagy, decreased mTOR protein levels have been held responsible which could result from therapies that target either mTOR directly or via the interrelated pathway activities of PI3K or AKT [75]. De-repression of autophagy during genotoxic treatment of cancers however, which include cisplatin or radiation treatment, might be attributed to the DNA-damage-regulated actions of p53. In this sense, the nuclear stress-activated transcription factor p53 can directly mediate autophagy (and apoptosis) via the damage-regulated autophagy modulator (DRAM) which represents a lysosomal protein that is able to advance different stages of autophagosome formation [76,77]. Recently, even the activity of microRNAs has been implicated in modulating drug resistance via regulation of ATG genes which underlines their crucial role in autophagy [78]. Nevertheless, evidence for molecular details of the direct autophagy-causing activities of other cancer therapeutics is lacking thus far. As an alternative explanation it has been suggested that by various treatment therapies, cancer cells could be specifically selected that already undergo high levels of autophagy which were named therapy refractory cancer stem cells. Thus, in different types of tumors following irradiation, higher levels of autophagy were detected which responded to inhibition of autophagy by reduced survival of breast, lung and cervical cancer cell lines [55].

4. Histone Deacetylase Inhibitors and Their Effector Mechanisms

Deregulated histone and non-histone deacetylation as a major epigenetic regulatory mechanism of gene transcription has been recognized as the underlying cause of transcriptional deregulation of tumor-related genes in many solid as well as hematological tumors [79,80,81]. By impeding crucial physiological cellular processes related to cell death regulation, cell proliferation, differentiation, senescence, migration and adhesion, malignant transformation of the cell is facilitated [82,83,84,85,86]. In order to support the cancer cell in resisting imbalanced growth-promoting and cell death signals, in recent years HDACi have been promoted as encouraging anticancer agents to interfere with epigenetically deregulated gene.
Expression caused by aberrant post-translational protein modifications. Similar to well-established epigenetic drugs, as for example DNA methyltransferase inhibitors, HDACi reverse histone acetyltransferase- and deacetylase-mediated acetylation of both, histone and non-histone proteins. Although HDACi-mediated cytotoxic responses are very pleiotropic, and seemingly depend on the type of tumor and used drug doses, specific re-occurring mechanisms are common for many HDACi that ultimately trigger tumor cell death [87,88,89] (Figure 1). Major effector mechanisms of HDACi are found in the ability to re-induce cell cycle arrest by p53-dependent or -independent induction of the cyclin-dependent kinase inhibitor p21CIP/WAF1 (p21). These effects induce cell differentiation in transformed cells by downregulating positive regulators of cell proliferation, such as c-MYC and c-SRC [90,91,92,93,94,95,96,97]. It is assumed that accumulated DNA damage in arrested tumor cells then promotes the activation of apoptosis or mitotic cell death as it cannot exit the cell cycle and finish mitosis [98,99,100,101]. This mechanism is furthermore underscored by the ability of HDACi to enhance the formation of ROS possibly due to the exclusive suppression of the natural scavenger thioredoxin (TRX)- an intracellular antioxidant- via upregulation of TRX-binding protein 2 (TBP2) in malignant cells [102,103,104]. Other HDACi anticancer mechanisms cause interference with stress response pathways of the endoplasmic reticulum, affect chaperone protein function leading to altered stability and expression of oncoproteins, or facilitate the accumulation of misfolded proteins [105]. In addition, HDACi-mediated de-repression of metastasis-related genes, influencing migration and invasion behavior of human tumor cell lines, as well as altered angiogenic inhibition, due to re-expressed pro- and anti-angiogenic genes, was documented [106,107,108,109,110,111,112,113,114,115]. Moreover, histone deacetylases (HDAC) have been involved in directing the expression of DNA damage-related response proteins and the modulation of histone deacetylation at sites of DNA damage to maintain genomic integrity; thus, only recently, HDACi administration has been found to interfere with a range of DNA repair processes in response to the induction of autophagy by genomic damage [79,116,117,118,119].
Key determinants of HDACi-triggered lethality are apoptosis and autophagy in a multitude of tumor cells; due to re-expression of silenced tumor suppressor genes and/or oncogenes, inhibitory signals for cellular growth and elevated cellular stress are (re-)activated. Accordingly, direct modulation of the acetylation pattern of transcription factors belonging to the class of non-histone proteins, can re-activate cell death-related signaling pathways in transformed cells (e.g., NF-κB, p53, and STATs) [85]. For example, HDACi treatment determines the acetylation status of the transcription factor p53 and thereby influences its mutual direct interaction with the MDM2 E3-ligase which regulates its degradation in H1299 carcinoma cells [120].

5. Resistance to HDAC Inhibitor-Induced Autophagy

Treatment with HDACi has exposed several varying molecular signaling mechanisms leading to the activation or suppression of HDACi-mediated autophagy (reviewed previously in [121,122,123,124,125,126,127] Figure 2 and Table 1). In by far most cases, mTOR inhibition involving the canonical pathway, followed by ROS accumulation, NF-κB hyperacetylation, p21 upregulation, or the involvement of p53 signaling, is observed during autophagic activation [49,58,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142]. Individual reports of HDACi-induced autophagy also detail apoptosome inactivation, increased transcriptional activity of FoxO1 (forkhead box O1) or NRF2 (Nuclear factor erythroid 2-related factor 2), upregulation of DAPK (death-associated protein kinase) and nuclear translocation of AIF (apoptosis inducing factor) [143,144,145,146,147]. Nevertheless, beside these defined mechanisms, direct HDAC-mediated acetylation of many ATG proteins, including ATG3, ATG7 or ULK1 (UNC-51-like kinase 1) has been brought in evidence, in addition, or as the only findings [85,148,149,150]. HDACi-mediated suppression of autophagy additionally involved mTOR attenuation in two reports [64,151,152].
HDACi-elicited autophagy has also been linked with the occurrence of therapeutic resistance [58,185,186,187,188]. Consequently, a discussion is ongoing, whether induction of autophagy stimulates antitumor activity or assists in the development of chemotherapeutic or radiotherapeutic resistance; the later might be true as malignant cells are exceedingly adaptable to inhibitory survival stimuli and cytotoxic agents [189,190]. As a result of permanent selection during tumor development and chemotherapy, particularly cancer cells are finally able to deal with a range of harmful conditions such as oxidative stress or DNA damage that renders them independent from external growth signals. In addition, stable genetic or epigenetic alterations of tumor cells that initially trigger tumorigenesis and particularly modify death signaling, might also be detrimental factors that inevitably determine the outcome of the autophagic response. Considering the increasing number of clinical trials performed the identification of presumable counteractions of potential drug combinations and the improvement of their efficacy gains significant weight.
This is particularly necessary, as only few in vivo data are available and research focused on only a few multidrug-resistant cell lines (linked to P-glycoprotein) [191,192,193]. Previous studies underscored molecular mechanisms, such as alterations in HDAC protein expression, retinoic acid signaling deregulation, and multidrug resistance facilitated by ATP-binding cassette transporters (MDR1 and its product, P-glycoprotein) as emanating cause of HDACi resistance [186,194,195,196]. As a predictive biomarker, constitutive STAT (signal transducers and activators of transcription) activation has been reported in SAHA (vorinostat)-treated CTCL patients [197]. Related to the identified triggering signals of HDACi-elicited autophagy, we summarize below the characterized and potential molecular basis of resistance mechanisms (Figure 2 and Table 1).

5.1. mTOR Modulation

The nutrient-sensing kinase mTOR (mammalian target of rapamycin) is a primary suppressive coordinator and upstream key component of the autophagic signaling machinery that regulates the ULK1 complex [3,4,198]. mTOR associates with unique binding partners and thereby reacts and controls, as a central interface of growth factor and nutrient signaling, many cellular functions via two multiprotein complexes, mTORC1 and mTORC2; this involves, for instance, proliferation, growth, survival, motility, and metabolism [199,200,201,202,203]. Due to the versatile modulation of the mTOR complexes, the stimulation and suppression of autophagy can be achieved in many ways. Thus, they are controlled on the one hand by PI3K (phosphatidylinositol 3-kinase)/AKT (a serine-threonine kinase encoded by the human homolog of the oncogene in the transforming retrovirus, AKT8; also called protein kinase B (PKB) but also by AMPK (AMP-activated kinase). AMPK acts as a central sensor for energy and metabolism (such as lipid, cholesterol and glucose metabolism) that is specifically controlled by Rag GTPases for amino acid availability, by REDD1 for oxygen availability, and by p53 for DNA damage in the cell [204,205,206,207]. During times of sufficient supplies, initiated by the phosphorylation of AKT, mTORC1 generally suppresses the activation of autophagy by phosphorylating itself the ULK1 complex further downstream, and only basal autophagy can take place [208,209]. During conditions of nutrient deprivation or shortage, the induction of autophagy is essential for cell survival, as cellular components can be reused and provide metabolic supply. In this event, AMPK can be activated by phosphorylation mediated by its potential main upstream regulator, the serine-threonine kinase LKB1 (liver kinase B1) that supports activation of the tumor suppressor tuberous sclerosis protein (TSC2) [210]. GSK3 (Glycogen synthase kinase-3)-initiated activation of the histone acetyltransferase TIP60 and consequent acetylation of ULK-1 represents a further possible mechanism of starvation-induced autophagy [211]. mTORC1, that can be inhibited by rapamycin, plays also a detrimental role in regulating protein translation by controlling the phosphorylation of the elongation factor 4E-BP1 and p70S6 kinase [212]. These two factors are presumed to mediate mTOR-induced suppression of autophagy [213,214].
As a further major downstream player in the canonical mTOR-activated autophagic signaling cascade-mediated by ULK1, Beclin-1 is required for the organization of autophagosome formation [160]. Beclin-1 enables autophagy mainly through forming extensive protein complexes with other autophagy-related proteins belonging to the Beclin-1 interactome (e.g., Vps34, p150, UVRAG, Bif1, Atg14L, Rubicon) [215]. Modifications of transcriptional regulation of pro-apoptotic (BAX, BAD, BNIP3, or PUMA), or anti-apoptotic members (BCL-2, BCL-XL, and MCL-1) of the BH3-only family, inhibit or activate Beclin-1-dependent autophagy by direct interaction with the BH3 domain of Beclin-1 [216,217,218,219].
The PI3K/AKT/mTOR signaling axis presents comparably one of the most frequented pathways of HDACi-activated autophagy that is often supported by increased ATG protein expression, such as LC3 (microtubule-associated protein 1A/1B-light chain 3) and Beclin-1 [49,135,136,137,138,139,151,153,154]. SAHA-mediated inactivation of mTOR, for example, induces autophagy by restoring the function of the ULK1 complex [134,136,137,138,139]. Initially, this undispensable role of mTOR in the regulation of SAHA-induced autophagy was experimentally verified by studies of our own group on uterine sarcoma cells and by Gammoh et al. [49,136,153]. Nevertheless, the ULK1 complex has an adequate function in mediating HDACi signaling, as ULK1-deficient cells have been documented to block SAHA-elicited autophagy [136]. Interestingly, two cases also reported about HDACi-elicited suppression of autophagy by activating the PI3K/AKT/mTOR pathway. In the first case, MGCD0103 enhanced the degradation of autophagy-related proteins in primary CLL (chronic leukocytic leukemia) cells via silencing ATG7 expression [151,152]. In the second report, accumulation of mitochondria and ROS, DNA damage, and apoptosis were detected in down syndrome-associated myeloid leukemia cells (DS-AMKL cells) that were treated with various HDACi (SAHA, TSA (trichostatin A), VPA (valproic acid), MS-275 (etinostat), JQ2) resulting in suppression of baseline autophagy due to activation of mTOR [64].
Considering the emanent role of the PI3K/Akt/mTOR signaling cascade in controlling essential cellular functions, it is not an unanticipated finding, that in many cancers dysregulation can be found in this pathway [220,221]. Thus, therapeutic resistance can be caused for instance by constitutive PI3K-AKT signaling that hyperactivates mTOR and therefore inhibits autophagy, which is encountered in many cancer cells; this is often due to mutations in PI3K itsself or other upstream signaling factors, loss of the tumor suppressor genes PTEN (phosphatase and tensin homolog) and ARHI, or by AKT overexpression [155,156,157,158]. Particularly, about 60% of breast cancer tumors accommodate genetic variations leading to high activity of the PI3K/AKT/mTOR signaling route which have been classified as oncogenic driver mutations [222]. Consequently, many targeted therapeutic interventions that address the reversal of endocrine resistance, apply inhibitors for mTOR (such as rapamycin (sirolimus) or its rapalogs temsirolimus, everolimus, deforolimus, etc.), for PI3K (buparlisib, pictilisib, copanlisib, alpelisib, and taselisib), and for AKT (ipatasertib) which entail diminished mTOR activity and therefore derepression of autophagy [223]. Due to off-target inhibition and adaptive autophagic resistance, it is not surprising that major challenges in developing optimized and rational drug combinations for the PI3K/AKT/mTOR signaling pathway persist.
Resistance to Beclin-1-mediated autophagy arises mainly due to monoallelic loss of the tumor suppressor protein Beclin-1, particularly in sporadic breast, ovary, and prostate tumors, or due to overexpression of the oncoprotein Bcl-2 related to an aggressive clinical tumor phenotype in a variety of cancers [160,161,162,163]. Thus, direct obstruction of Bcl-2 mediated autophagic cell death was documented in breast cancer cells which could be overcome by gene silencing, thereby offering a novel therapeutic strategy [218,224].

5.2. ROS Accumulation and Enhanced Antioxidant Expression

ROS are induced by various kinds of cellular stress and disturb the mitochondrial respiratory chain and energy metabolism. Due to this activity, they have frequently also been involved in ROS-induced dual activation of apoptosis and autophagy [225]. Here, selective autophagy in the form of mitophagy-regulated by NIX/BNIP3L- and PINK1-mediated pathways represents a tumor suppressor mechanism, as the elimination of damaged mitochondria prevents the further accumulation of ROS [226,227,228]. Mitochondrial damage and endoplasmatic reticulum stress resulting in the formation of ROS might furthermore lead to an autophagic response via activation of transcription factors such as FOXO and ATF4 that in turn increase the expression of ATG genes (LC3, ATG5) [229,230,231]. A further mechanism of ROS-induced autophagy could be mediated by Ataxia-telangiectasia mutated (ATM), a cellular damage sensor that coordinates the cell cycle, with damage-response checkpoints and DNA repair, to preserve genomic integrity [232]. In response to elevated ROS, ATM activates TSC2, via the LKB1/AMPK metabolic pathway in the cytoplasm, to repress mTORC1 and induce autophagy.
ROS induction represents a further common factor that initiates the activation of HDACi-mediated autophagy in tumor cells [233]. It has been reported to occur as a single event or in combination with mTOR deactivation, due to extensive intracellular ROS generation [122,123]. Upon deleting ATG5 or ATG7 in cells, the use of autophagy inhibitors in the presence of ROS has been demonstrated to promote tumor development, due to the culmination of deleterious mitochondria and chronic oxidative stress as well as tissue damage and inflammation [234,235,236].
The specific mechanism leading to HDACi-induced accumulation of ROS has not been resolved up to date but seem to lie in the posttranslational modification of TRX, representing an intracellular antioxidant that serves as a natural scavenger of ROS. Corresponding evidence was derived from the finding that increased TBP-2 expression could be localized in normal but not in malignant fibroblasts in humans which decreases the reductive capacity of TRX [102]. Thus, tumor cells treated with HDACi have a lowered ability to handle oxidative injury. This assumption is consistent with the analysis of several tumors where, in addition to ROS generation, activation of the MAPK (mitogen activated protein kinase) family members, ERK1/2 and JNK or elevated expression of the lysosomal protease cathepsin D in combination with diminished levels of its substrate, TRX could be detected [58,137]. In the case of MS-275 treated human colon cancer cells, p38 MAPK even played a vital role in switching between HDACi-mediated apoptosis and autophagy [166,237]. In one report however, HDACi (VPA, TSA)-mediated autophagy and cytotoxicty occurred in pancreatic cancer cells obviously due to concomitant increased ROS production and HDACi-induced ERK1/2 inhibition which promoted c-MYC downregulation, a survival factor for cancer cells exhibiting constitutively activated RAS [130]. This depicts the upregulation of enzymes related to cellular redox control, anti-oxidative stress, and energy metabolism in the proteomic study of SAHA-induced Jurkat T-leukemia cells [137]. Contrastingly, ROS-mediated autophagy promoted survival of AML1-ETO cells in response to SAHA and VPA treatment due to the removal of ubiquitinated proteins; this could be overcome by the use of HDACi and CQ cotreatment; dependency on ROS for the mediation of cell death could be clearly demonstrated by the use of the ROS scavenger N-acetylcysteine (NAC) in these cells [62].
Several anticancer therapies that stimulate ROS- induced autophagy and possibly apoptosis have also been linked with the development of cell death resistance [44,59]. As many cancer targeting drugs also promote cellular stress, the increased formation of ROS could also upregulate tumor cell-protective levels of autophagy that facilitate cancer cell survival. In the case of ROS-induced autophagic resistance, one could therefore also postulate that some classes of HDACi might not exert a perfect suppressive effect on TRX expression levels; this might be particularly relevant for cancer cells where apoptosis is not activated in parallel by the used HDACi. Accordingly, increased levels of ROS scavenging and anti-oxidant enzymes have been detected in multidrug-resistant cancer cells [164]. In this case, it is hard to predict whether the use of autophagy inhibitors might restore the sensitivity to the treatment thereby modulating cancer cell proliferation, survival, and drug resistance.

5.3. p21CIP/WAF-1 Upregulation

Many HDACi exert antiproliferative effects by re-inducing arrest in the G1 or G2 phase of the cell cyle and by inducing differentiation through upregulating p21 expression [93,238]. The consequent cyclin-mediated inability to pass two cell-cycle checkpoints is presumed to exert a protective function on the transformed cell allowing its repair. In the case of accumulated DNA damage in tumor cells (such as DNA double-strand breaks), the inability to further proceed in the cell cycle causes an early escape from an unfinished mitosis and the subsequent induction of cell death [95,98,100,101,239]. CDKN1A, the gene enoding for p21, is one of the most prominent target genes of p53, nevertheless, also p53-independet induction of p21 has been reported. Acetylation and HDAC1-mediated deacetylation of p53 have been shown to mutually regulate binding of p53 to the promoter region of CDKN1A, and its expression [238,240]. p53 is also the main regulator of p53R2, a human ribonucleotide reductase homologue that contains a p53-binding site in intron 1 which up-regulates p21 [241].
HDACi-mediated upregulation of p21 can promote cell death via apoptosis, and in two reported cases also via autophagy, but the underlying mechanism is poorly resolved. In the first case, p21-mediated autophagy occurred in addition to cell differentiation and cell cycle arrest; this was mediated by the HDACi combination of H40 and SAHA resulting in hyperacetylation of histone H3 and upregulated p21 expression in PC-3M and HL-60 cells [142]. In the second event, the HDACi MRJF4 was found to induce p21-elicited autophagy in PC3 cells [128]. In both studies, additional downregulation of NF-κB signaling was also documented, which could be assumed as the primary cause of autophagic induction. In a further study of apicidin-induced autophagy in human oral squameous cell carcinoma cells, p21-mediated G2/M phase arrest was accompanied by mTOR attenuation [134,159]. In two reports of uterine and pancreatic cancer cell autophagy, concomitant with either p53-deficiency or HDACi-induced mutant p53 induction, upregulation of its targets p21 could be observed [49,130].
Cytoplasmic localization of p21, possibly due to deregulated Akt1-mediated phosphorylation or truncation of the nuclear localization signals has been related with an anti-apoptotic gain-of-function [172,173]. The anti-apoptotic function of p21 includes inhibition of caspases, such as CASP3, 8, and 10, stress-activated protein kinases (SAPKs), and apoptosis signal-regulating kinase 1 but can also involve the up-regulation of anti-apoptotic protein encoding genes [172,242]. As cytoplasmic p21 has been frequently identified in aggressive tumors that are particularly associated with poor prognosis, this finding could nevertheless also represent a cause of autophagic resistance [174]. Deregulation of Akt can thereby occur through genetic modifications of PI3K, but also by loss of PTEN, or by HER2/neu (ERBB2) amplification. Moreover, p53-mediated upregulation of p21 via p53R2, also targets cytoplasmic p21 and thereby facilitates the progression of cancer [241]. Consistently, p53 and p53R2 may have a tumor-promoting function in cancers with cytoplasmic p21 (and Akt overexpressing cancers), and support the development of chemoresistance against anticancer therapeutics leading to poor prognosis. Furthermore, radioresistance via DNA damaging therapies, such as UV irradiation, can be obtained in this way as the p53R2-p21 interaction is weakened and levels of non-bound nuclear p21 increases [175]. Consequently, also various forms of mutated or deficient p53 and its regulatory proteins are able to cause p53R2-mediated deregulation of p21 and the formation of resistance against HDACi-induced cell death.

5.4. p53 Deficiency and Acetylation

The initial identification of p53 as an important regulator of autophagy was established by the clarification of its target gene DRAM1 (damage-regulated autophagy modulator). DRAM not only directly activates apoptosis, but also autophagy, when DNA-damaging agents induce p53 transactivation [76]. It represents a lysosomal protein that is entangled in different phases of autophagosome formation and could also be activated by the p53 relative, p73 [77,243]. Positive transactivation-dependent regulation of autophagy involves furthermore p53-mediated activation of the tumor suppressor genes genes TSC2 (tuberous sclerosis complex 2) and PTEN (phosphatase and tensin homolog), as well as AMPK, or its activators sestrins 1 and 2 [157,244,245,246]. The further signaling cascade of these pro-autophagic factors then follows the canonical pathway of autophagy, mediated by mTOR attenuation. p53 can furthermore influence the expression of pro-apoptotic proteins (downregulation of BCL-2, BCL-xL, and MCL-1), anti-apoptotic proteins (upregulating BAX, BAD, BNIP3, or PUMA), or the tumor suppressor protein p14ARF (alternate reading frame protein product of the CDKN2A locus) leading to the release of Beclin-1 [218,247,248]. p14ARF seems to be important in stabilizing the p53 protein and maintaining its cell death regulation ability, in order to oppose hyperproliferative signals causing oncogenic activation [249,250]. As a further p53-regulated protein, DAPK induces autophagy, either by phosphorylating and stabilizing Beclin-1 against BCL-2/BCL-xL-mediated degradation, or by blocking the anti-autophagic activity of the LC3-interacting MAP1B protein [217,251,252]. Negative regulation via p53 protein, located in the cytoplasm, has been additionally uncovered at last [253,254,255,256,257]. Although the exact mechanism still needs to be elucidated, this inhibitory function is a transcription-independent process that finally also targets the identical AMPK-mTOR signaling cascade used by nuclear p53 transactivation-dependent autophagy. Under normal physiological conditions, in contrast to nuclear p53 activity, direct interaction of p53 protein with AMPK increases mTOR activity, leading to autophagic suppression [256]. This tumor-suppressive mechanism, resulting in increased autophagic flux in p53-deficient cells, is presumably intended to provide elevated resistance to metabolic stress. mTOR-independent inhibition of autophagy by cytoplasmic p53 has been furthermore shown to be mediated by TIGAR (TP53-induced glycolysis and apoptosis regulator) resulting in downmodulation of glycolysis and of ROS formation, as a response to stress [258].
HDACi-dependent autophagic induction involving p53 was previously reported by our own study, that revealed p53 deficiency as the cause of mTOR signaling-mediated autophagy in SAHA-treated ESS-1 uterine sarcoma cells, and in MDA-MB-231 breast cancer cells by Fogetti et al. [49,122,123,237,259]. Similarly, to our finding, the HDACi VPA and TSA provoked, in response to p53 deficiency, concomitant upregulation of apoptosis and autophagy in pancreatic cancer cells [130]. However, opposed to homozygous inactivation of TP53 in ESS-1, cell death activation was mediated by reduced mutant p53 expression in this case due to ERK-mediated stabilization of the oncogenic protein c-MYC, and by reactivation of wild-type p53 expression. Increased acetylation of the p53 protein following specific inhibition of class III NAD-dependent deacetylases SIRT1 and SIRT2 by sirtinol or combined sirtinol/MHY2256 treatment, preventing MDM2-mediated degradation, yielded elevated LC3-II expression and autophagy, in addition to cell cycle arrest and apoptosis in MCF-7 breast cancer cells or in endometrial cancer cells, respectively [131,132,133].
While intact p53 protein is essential for maintaining stress-mediated responses and DNA repair in the cell, impairment of its integrity by mutation is frequently found in almost all types of human cancers [260]. Altered p53 functions, most commonly due to missense mutations, favor malignancy and resistance to chemotherapy. These genetic alterations lead to a loss in the trans-activation capability of the transcription factor rendering the cell vulnerable against applied stress and malignant transformation; alternatively, they generate dependence on alternative pathways or proteins required for cell survival. Depleting these redundant survival pathways in p53-deficient cells is therefore one of the objectives of future cancer treatment. Overly resistance to chemotherapeutic drugs, including cisplatin, alkylating agents, anthracyclines, antimetabolites etc., was often related to overexpression of mutant p53 that possesses conformational alterations. Thus, mutant p53-reactivating small molecule compounds such as CP-31398, WR-1065, PRIMA-1, and MIRA-1, or the blockage of proteasomal degradation might help to restore p53-dependent cell death and tumor suppression [261,262,263,264,265,266]. Frequently, however, these heterozygous mutations acquire, due to hyperstabilization and accumulation, dominant-negative or gain-of-function activity over p53 protein, derived from the intact allele (e.g., by impeding access to the promoter), which finally confers pro-ongogenic roles; these range from malignant cell proliferation and invasion to the development of drug resistance [167,168,169]. Additionally, or due to these gain-of-function mutations, p53 activity may be also be defective caused by alterations in regulatory proteins of p53, particularly of MDM2 that determines its half life. Reduced mutant p53 degradation is therefore facilitated by overexpressed short isoforms of MDM2 or auxillary chaperone proteins (such as HSP90, BAG family proteins) [170,171].
As one of the effector mechanisms, it was elucidated that the interaction of HDACi with mutant p53 protein enables upregulation of p21 and MDM2, and thus its proteasomal degradation [267,268,269]. This could be demonstrated by treating heterozygous p53-deficient cells with TSA, FR901228, or SAHA, which re-established p53-dependent transcription, either by re-activating wild-type expression of p53 itself, by copying p53 transcriptional activity, or by upregulating autophagy [130,268,270]. In a further report, release of mutant p53, by SAHA-facilitated disruption of a complex consisting of HDAC6 and heat shock protein 90 (HSP90), enabled MDM2 and CHIP ligase-mediated degradation [269]. Additionally, the transcription factor HOXA5 was documented to upregulate mutant p53 mRNA and protein expression in tumor cells following SAHA, NaB (sodium butyrate) or HDAC8 administration [271]. Very recently, HDACi-mediated degradation of mutant p53, through the activation of autophagy, has also been reported. In the human breast cancer lines MDA-MB-231 (mutp53-R280K) SAHA induced significant depletion of mutant p53 supporting tumor survival that could be blocked by inhibition of autophagy leading to increased cell death [259]. Also in our previously described model of the uterine sarcoma cell line ESS-1, the activation of SAHA-induced autophagy could be explained by elimination of mutant p53 (mutp53-R213X) and the lack of p53 protein; although not yet experimentally verified, which could target the cytoplasmic p53-mediated negative regulatory pathway of autophagy [49]. These findings support the anticancer strategy of engaging cellular pathways that facilitate mutant p53 degradation which may not only reduce the oncogenic potential of cell growth and invasion but also increase the sensitivity to anticancer drugs. Accordingly, beside HDACi, DA several encouraging HSP90 inhibitors (such as 17-AAG or ganetespid) and MDM2 inhibitors (nutlins, benzodiazepinediones or spiro-oxindoles) have been proposed that affect the p53-MDM2 signaling axis, and therefore open new possibilities for the development of more efficient anticancer drugs [272,273].

5.5. NF-κB Hyperacetylation

In addition to their well-known role in regulating immune responses, the NF-κB (Nuclear factor kappa B) transcription factor members (RelA, RelB, c-Rel, NF-κB1, and NF-κB2) are critical anti-apoptotic factors that sustain cell growth and survival and are frequrently deregulated in cancer. Under normal conditions, proteins of the NF-κB family are kept inactive by several inhibitory cytoplasmic proteins belonging to the IκB kinase complex [274,275]. In tumor cell lines, NF-κB has been furthermore established as a suppressive regulator of autophagy, in response to the cytokine TNFα, which may contribute to suppression of TNFα-induced apoptotic signaling and oxidative stress. Following the induction of NF-κB mediated autophagy, the autophagy-associated proteins Beclin-1 and p62/SQSTM1 were found upregulated; IKK kinase in contrast, an activator of NF-κB was degraded by autophagy, due to a loss of Hsp90 function, leading to the inhibition of NF-κB signaling. The exact mechanisms of autophagic induction are unknown but could involve mTOR signaling or interference with Beclin-1/Bcl-2 stabilization. Conversely, inhibition of NF-κB signaling was reported to augment starvation-induced autophagy [276]. Interestingly, TSC2, an inhibitor of the mTOR pathway, positively regulated NFkB activity revealing a reciprocal relationship between NF-κB and autophagy that might represent a feedback loop of cell death signaling.
NF-κB-mediated autophagy induced by HDACi has been reported in three studies. Combined SAHA and MS-275 treatment of PC3 cells positively regulated autophagy by by NF-κB RELA/p65 hyperacetylation leading to transcription of several NF-κB target genes and suppressed the innate immune system in vesicular stomatitis virus oncolysis [141]. In two reports, induction of autophagy was preceded by suppression of pERK/NF-κB signaling and upregulation of p21 in PC-3M (and HL-60) cells [128,142].
NF-κB and its activating kinase, IKK, have attracted therapeutic intervention because of their significant function in the progression, not only of cancer, but also of many other diseases involving chronic inflammation. Besides the regulation of cell death, NF-κB signaling has critical immunomodulatory roles in arranging cytokine release, regulating prostaglandins and redox reactions, and in governing angiogenesis [277]. NF-κB upregulation or constitutive active signaling, in cancer or normal cells, suppresses apoptosis, wheras it promotes angiogenesis and metastasis thereby causing resistance to radiotherapy and chemotherapy which reduces therapeutic efficiency. Nevertheless, as many other pathways convene at the NF-κB pathway, including those of other immune-chemotherapeutic triggered phenotypes of resistance, it is hard to determine the specific molecular culprit [176]. Either the application of selective inhibitors, or the use of natural NF-κB agents (such as curcumin, melatonin, resveratrol, and quercetin), have shown encouraging results for tumor sensitization but need to be verified in clinical studies.

5.6. FOXO1 Transcription

FOXO1 is a member of the FOXO family that serve as transcriptional regulators in many intracellular processes, such as cell growth, proliferation, differentiation, and longevity, including regulation of autophagy [278]. Via post-translational modification of FOXO1, the modulation of distinct sets of target genes can be activated that execute functions in silencing oxidative stress by regulating anti-oxidative enzymes, metabolic and immune effectors, DNA repair, cell cycle arrest and apoptosis, as well as autophagy-associated genes [279,280,281]. Involved triggers of FOXO1 activity include oxidative stress, high glucose, and several more, that are mediated by PI3K-AKT, JNK, CBP, sirtuins (SIRT), or ubiquitin E3 ligases signalling pathways [282]. FOXO1-driven autophagy or apoptosis has been demonstrated as an important function in response to stress; it is regulated by acetylation of FOXO1 following its dissociation from SIRT2—a mainly cytoplasm-located deacetylase- and by subsequent binding to ATG7, a key regulator in the formation of the autophagosome [245]. But acetylation of FOXO1 and activation of autophagy can also be triggered by the natural agents benzyl isothiocyanate and curcumin, as well as by starvation [283,284].
FOXO1-stimulated transcriptional activation of SAHA and TSA-treated HepG2 and HCT116 cells was also declared as a mediator of HDACi-induced autophagy [145]. As underlying mechanism, sestrin 3 (SESN3)-mediated suppression of mTOR and TSC2, in addition to upregulation of ATG gene expression (such as ATG4B, ATG12, PIK3C3, BECLIN-1, and MAP1LC3B), could be detected. Due to their commonly altered expression in many tumors, cytosolic FoxO1-modulated cell death is considered as tumor suppressor activity. Regulatory pathways involved in the oncogenic activity of FOXO factors have been including PI3K, Ras, IKK signaling but also microRNAs and have been linked to the deregulation of cell cycle arrest (p27KIP1, p21) and cell death genes (e.g., FASL, TRAIL, BIM) [285,286].
Nevertheless, recent reports elucidate a more intricated role of FOXO factors by contributing to cancer proliferation and resistance to treatment. Mechanisms of resistance have been found, on the one hand in elevated expression of the multidrug resistance protein 1 (MDR1) in breast cancer cells in response to to adriamycin, or on the other hand by neutralizing the effect of oxidative-stress promoting agents due to FOXO1-elicited upregulation of anti-oxidative enzymes (e.g., paclitaxel-resistant ovarian cancer) [177,178,179]. A further revealed cause of drug resistance was presented in renal cell carcinoma that was provoked by inhibitory drugs of the PI3K pathway. Subsequent AKT phosphorylation and activation causing renal tumor growth was mediated by upregulated FOXO activity and mTORC2 induction (via RICTOR expression) in an amplifying loop [287].

5.7. DAPK Upregulation

DAPK is a calcium/calmodulin-regulated protein kinase that is involved in the regulation of various cellular processes, such as caspase-independent cell death, apoptosis, and cytoskeletal activities [288]. It furthermore induces cell death signaling in response to inflammatory stimuli, such as interferon-gamma (INF-γ), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta (TGF-β) where it interacts with different MAPKs, such as ERK [289]. DAPK has also been reported as an important regulator of autophagy involving several mechanisms [290]. By modulating the Vps34 class III PI3K complex, DAPK interferes with the process of autophagosome nucleation. This invovlves either DAPK-mediated phosphorylation of protein kinase D, which in turn phosphorylates Vps34, or the phosphorylation of Beclin-1, that also represents an essential component of the Vps34 complex [217,291]. Phosphorylation of of the BH3 domain of Beclin-1 by DAPK causes it to dissociate from anti-apoptotic BCL-2 family members such as Bcl-2 or Bcl-XL, and this event promotes the inhibition of the PI3K complex and stimulation of autophagy [218,251]. Moreover, independently from Beclin-1, DAPK has also been shown to activate autophagy via interaction with LC3 [252].
LBH589/panobinostat-mediated dephosphorylation of serin308 of DAPK affecting direct protein interactions was determined as the single cause of DAPK-induced autophagy in HCT116 colon cancer cells [147].
DAPK-based drug resistance against erlotin and cetuximab in cell lines of non-small cell lung cancer or head and neck squamous cell carcinoma was found to be caused by hypermethylation of the DAPK-encoding gene [180]; this could be reproduced by siRNA-mediated knockdown of DAPK while a gene transfers rescued resistance. Similarly, TSA re-sensitized cisplatin-resistant A549 cells to apoptosis by up-regulating DAPK [292]. Another study identified KLHL20-Cul3-ROC1 as an E3 ubiquitin ligase for DAPK-mediated degradation and therefore a regulatory mechanism that could explain resistance to INF-γ cell death signaling in cancer therapy [181].

5.8. NRF2 Upregulation

NRF2 is a transcriptional regulator of oxidative as well as xenobiotic stress (environmental toxicants or harmful chemicals)-induced cytoprotective genes, such as glutathione peroxidase, superoxide dismutase, and thioredoxin. Corresponding signaling is mediated by the NRF2-KEAP1 (kelch-like-associated protein 1)-Antioxidant Response Element (ARE) pathway. Autophagy has been implicated in the regulation of oxidative stress by undergoing the NRF2/KEAP1) and SQSTM1/p62 pathway [293,294,295]. SQSTM1/p62 represents an autophagy cargo adapter and substrate that can bind and guide KEAP1 to the autophagosome, thereby enabling its selective degradation [296,297]. This interaction furthermore releases NRF2 which subsequently translocates to the cell nucleus, where it binds to ARE sequences, which transcriptionally activates the expression of about 500 different genes, many of them possessing antioxidant effects [298]. In contrast to physiological conditions, where NRF2 is sequestered by KEAP1 and inactivated by proteasomal degradation, oxidative stress prolongs the extension of its half-life and transcriptional regulation involving, either upregulated SQSTM1/p62, or inhibited KEAP1 activity.
NRF2-signaling, mediated by microRNA-mediated regulation of mTOR, was also delineated as a novel regulator of HDACi-induced autophagy [146]. Thus, treatment of the hepatocyte-derived carcinoma lell line Huh-7, and of the primary gastric adenocarcinoma cell MGC-803, with TSA and SAHA upregulated NRF2 which subsequently increased expression the miRNA-129-3p by binding to its ARE1 site. MiRNA-129-3p in turn, induced autophagy and presumably also cell death by inhibiting mTOR, increasing the conversion of LC3-I to LC3-II, and downregulating SQSTM1/p63 expression.
Comparable to other autophagy-associated genes, aberrant expression of NRF2, due to reduced KEAP1 activity, is correlated to chemoresistance by facilitating stress reduction [182,183,184]. In fact, NRF2-activated autophagy has been identified as an underlying mechanism. Consequently, a knock-down of NRF2 yielded increased sensitivity of tumor cells to anticancer agents; moreover, cancer patients with low NRF2 levels had better results following chemotherapeutic treatment [299,300]. Thus, the NRF2-miRNA-129-3p-mTOR signaling axis, that represents a sensor for chemical-induced oxidative stress, is important for the maintenance of cellular homeostasis; it has been potentially associated with drug resistance and might serve as a useful therapeutic target [301].

6. Concluding Remarks

Chemotherapeutic resistance has been considered a major challenge in cancer that also affects HDACi treatment [185,186,188]. While the development of isoform-selective HDAC-specific inhibitors are still in progress, many lines of evidence including experimental and clinical studies attest a promising effect for the use of HDACi as epigenetic drugs in cancer treatment; nevertheless, several obstacles related to solid malignancies have been encountered [302,303,304]. Furthermore, exact mechanistic insights related to the effector and resistance mechanisms of HDACi-induced cell death and the evaluation of molecular determinants, enabling a reliable indication of the treatment outcome, are still incomplete. This might be owned to the non-exhaustively explored, versatile mechanisms of action of HDACi, influenced by the class and dose of HDACi, and their combinatiorial effects with other drugs on the one hand. Nonetheless, reasons could also be found in cancer-cell specific effects and context-dependent molecular effects influenced by tumor-specific mutations or microenvironmental conditions (e.g., oxidative stress), on the other hand.
Autophagy is a potential target for cancer therapy but is associated with an ambiguous role in diverse phases of tumorigenesis. Consistently, HDACi-mediated autophagy has been attributed with a critical role in HDACi resistance. Particularly, as the combination of inhibitors of autophagy with HDACi treatment has been established as an advanced therapeutic approach to re-sensitize cancer cells, the definition of decisive factors that direct autophagy towards a tumor suppressive or survival mode would be essential [47,187]. Comparable to apoptosis however, autophagy is governed by an intriguingly complex network of cellular signaling pathways. A focus of future studies should therefore pose the clarification of additional, non-elucidated molecular mechanisms of HDACi-mediated cell death and its determinants of resistance. The acquired information will allow the design of improved anti-tumor HDACi-composed drugs that include targeting of autophagy and even enable the selection of cancer therapy-responsive beneficiaries prior to treatment.

Author Contributions

M.M. and L.F.F. were equally responsible for conceptualization, writing—original draft preparation, writing—review and editing, and visualization of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mizushima, N.; Noda, T.; Yoshimori, T.; Tanaka, Y.; Ishii, T.; George, M.D.; Klionsky, D.J.; Ohsumi, M.; Ohsumi, Y. A protein conjugation system essential for autophagy. Nature 1998, 395, 395–398. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, Z.; Klionsky, D.J. Mammalian autophagy: core molecular machinery and signaling regulation. Curr. Opin. Cell Biol. 2010, 22, 124–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Klionsky, D.J.; Emr, S.D. Autophagy as a regulated pathway of cellular degradation. Science 2000, 290, 1717–1721. [Google Scholar] [CrossRef] [PubMed]
  4. Jung, C.H.; Ro, S.-H.; Cao, J.; Otto, N.M.; Kim, D.-H. mTOR regulation of autophagy. FEBS Lett. 2010, 584, 1287–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dall, K.; Færgemann, N. Metabolic regulation of lifespan from a C. elegans perspective. Genes Nutr. 2019, 14, 25. [Google Scholar] [CrossRef] [PubMed]
  6. Mizushima, N.; Levine, B. Autophagy in mammalian development and differentiation. Nat. Cell Biol. 2010, 12, 823–830. [Google Scholar] [CrossRef] [PubMed]
  7. Maiuri, M.C.; Kroemer, G. Autophagy in stress and disease. Cell Death Differ. 2015, 22, 365–366. [Google Scholar] [CrossRef] [Green Version]
  8. Li, W.W.; Li, J.; Bao, J. Microautophagy: lesser-known self-eating. Cell. Mol. Life Sci. 2012, 69, 1125–1136. [Google Scholar] [CrossRef]
  9. Kaushik, S.; Cuervo, A.M. Chaperone-mediated autophagy: A unique way to enter the lysosome world. Trends Cell Biol. 2012, 22, 305–309. [Google Scholar] [CrossRef] [Green Version]
  10. Yang, Z.; Klionsky, D. Eaten alive: a history of macroautophagy. Nat. Cell Biol. 2010, 12, 814–822. [Google Scholar] [CrossRef] [Green Version]
  11. Klionsky, D.J.; Abdalla, F.C.; Abeliovich, H.; Abraham, R.T.; Acevedo-Arozena, A.; Adeli, K.; Agholme, L.; Agnello, M.; Agostinis, P.; Aguirre-Ghiso, J.A.; et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012, 8, 445–544. [Google Scholar] [CrossRef] [PubMed]
  12. Takeshige, K.; Baba, M.; Tsuboi, S.; Noda, T.; Ohsumi, Y. Autophagy in yeast demonstrated with proteinase-deficient mutants and conditions for its induction. J. Cell Biol. 1992, 119, 301–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Harding, T.; Morano, K.; Scott, S.; Klionsky, D. Isolation and characterization of yeast mutants in the cytoplasm to vacuole protein targeting pathway. J. Cell Biol. 1995, 131, 591–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Klionsky, D.J. Autophagy revisited: a conversation with Christian de Duve. Autophagy 2008, 4, 740–743. [Google Scholar] [CrossRef] [Green Version]
  15. Mizushima, N.; Komatsu, M. Autophagy: renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [Green Version]
  16. Mizushima, N.; Yamamoto, A.; Matsui, M.; Yoshimori, T.; Ohsumi, Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol. Biol. Cell 2004, 15, 1101–1111. [Google Scholar] [CrossRef]
  17. Mizushima, N.; Levine, B.; Cuervo, A.M.; Klionsky, D.J. Autophagy fights disease through cellular self-digestion. Nature 2008, 451, 1069–1075. [Google Scholar] [CrossRef] [Green Version]
  18. Kraft, C.; Martens, S. Mechanisms and regulation of autophagosome formation. Curr. Opin. Cell Biol. 2012, 24, 496–501. [Google Scholar] [CrossRef]
  19. Reggiori, F.; Ungermann, C. Autophagosome maturation and fusion. J. Mol. Biol. 2017, 429, 486–496. [Google Scholar] [CrossRef]
  20. Galluzzi, L.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Manuel, J.; Pedro, B.-S.; Cecconi, F.; Choi, A.M.; Chu, C.T.; Codogno, P.; et al. Molecular definitions of autophagy and related processes. EMBO J. 2017, 36, 1811–1836. [Google Scholar] [CrossRef]
  21. Yang, K.; Sathiyaseelan, P.; Ho, C.; Gorski, S. Evolution of tools and methods for monitoring autophagic flux in mammalian cells. Biochem. Soc. Trans. 2018, 46, 97–110. [Google Scholar] [CrossRef] [PubMed]
  22. Rubinsztein, D.C.; Shpilka, T.; Elazar, Z. Mechanisms of autophagosome biogenesis. Curr. Biol. 2012, 22, R29–R34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Axe, E.; Walker, S.; Manifava, M.; Chandra, P.; Roderick, H.L.; Habermann, A.; Griffiths, G.; Ktistakis, N. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J. Cell Biol. 2008, 182, 685–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Shibutani, S.; Yoshimori, T. A current perspective of autophagosome biogenesis. Cell Res. 2014, 24, 58–68. [Google Scholar] [CrossRef]
  25. Mizushima, N.; Yoshimori, T.; Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu. Rev. Cell Dev. Biol. 2011, 27, 107–132. [Google Scholar] [CrossRef]
  26. Yanxiang Guo, J.; White, E. Autophagy, Metabolism, and Cancer. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 73–78. [Google Scholar] [CrossRef]
  27. Levine, B.; Mizushima, N.; Virgin, H.W. Autophagy in immunity and inflammation. Nature 2011, 469, 323–335. [Google Scholar] [CrossRef]
  28. Qu, X.; Yu, J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen, E.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 2003, 112, 1809–1820. [Google Scholar] [CrossRef] [Green Version]
  29. Eskelinen, E. The dual role of autophagy in cancer. Curr. Opin. Pharmacol. 2011, 11, 294–300. [Google Scholar] [CrossRef]
  30. Kubisch, J.; Türei, D.; Földvári-Nagy, L.; Dunai, Z.A.; Zsákai, L.; Varga, M.; Vellai, T.; Csermely, P.; Korcsmáros, T. Complex regulation of autophagy in cancer-integrated approaches to discover the networks that hold a double-edged sword. Semin. Cancer Biol. 2013, 23, 252–261. [Google Scholar] [CrossRef]
  31. Levy, J.M.M.; Towers, C.G.; Thorburn, A.; Foundation, A.; Brain, P.; States, U. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef]
  32. Shintani, T.; Klionsky, D.J. Autophagy in health and disease: a double-edged sword. Science 2004, 306, 990–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lorin, S.; Hamaï, A.; Mehrpour, M.; Codogno, P. Autophagy regulation and its role in cancer. Semin. Cancer Biol. 2013, 23, 361–379. [Google Scholar] [CrossRef] [PubMed]
  34. Morselli, E.; Galluzzi, L.; Kepp, O.; Vicencio, J.M.; Criollo, A.; Maiuri, M.C.; Kroemer, G. Anti- and pro-tumor functions of autophagy. Biochim. Biophys. Acta Mol. Cell Res. 2009. [Google Scholar] [CrossRef] [Green Version]
  35. Yang, Z.J.; Chee, C.E.; Hunag, S.; Sinicrope, F.A. The role of autophagy in cancer: therapeutic implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  36. Choi, A.M.; Ryter, S.W.; Levine, B. Autophagy in human health and disease. N. Engl. J. Med. 2013, 368, 651–662. [Google Scholar] [CrossRef]
  37. Rosenfeldt, M.T.; Ryan, K.M. The role of autophagy in tumour development and cancer therapy. Expert Rev. Mol. Med. 2009, 11, e36. [Google Scholar] [CrossRef] [Green Version]
  38. Morselli, E.; Galluzi, L.; Kepp, O.; Marino, G.; Michaud, M.; Vitale, I.; Maiuri, M.C.; Kroemer, G. Oncosuppressive functions of autophagy. Antioxdants Redox Signal. 2011, 14, 2251–2269. [Google Scholar] [CrossRef]
  39. Mathew, R.; Karantza-Wadsworth, V.; White, E. Role of autophagy in cancer. Nat. Rev. Cancer 2007, 7, 961–967. [Google Scholar] [CrossRef]
  40. Maiuri, M.C.; Tasdemir, E.; Criollo, A.; Morselli, E.; Vicencio, J.M.; Carnuccio, R.; Kroemer, G. Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ. 2009, 16, 87–93. [Google Scholar] [CrossRef] [Green Version]
  41. Su, Z.; Yang, Z.; Xu, Y.; Chen, Y.; Yu, Q. MicroRNAs in apoptosis, autophagy and necroptosis. Oncotarget 2015, 6, 8474–8490. [Google Scholar] [CrossRef] [Green Version]
  42. Lin, L.; Baehrecke, E.H. Autophagy, cell death, and cancer. Mol. Cell. Oncol. 2015, 2, e985913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yu, L.; Wan, F.; Dutta, S.; Welsh, S.; Liu, Z.; Freundt, E.; Baehrecke, E.L.M. Autophagic programmed cell death by selective catalase degradation. Proc. Natl. Acad. Sci. USA 2006, 103, 4952–4957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Poillet-Perez, L.; Despouy, G.; Delage-Mourroux, R.; Boyer-Guittaut, M. Interplay between ROS and autophagy in cancer cells, from tumor initiation to cancer therapy. Redox Biol. 2015, 4, 184–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Yue, Z.; Jin, S.; Yang, C.; Levine, A.J.; Heintz, N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA 2003, 100, 15077–15082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Mowers, E.E.; Sharifi, M.N.; Macleod, K.F. Autophagy in cancer metastasis. Oncogene 2017, 36, 1619–1630. [Google Scholar] [CrossRef]
  47. Lopez, G.; Torres, K.; Lev, D. Autophagy blockade enhances HDAC inhibitors’ pro-apoptotic effects: Potential implications for the treatment of a therapeutic-resistant malignancy. Autophagy 2011, 7, 40–41. [Google Scholar] [CrossRef] [Green Version]
  48. Han, J.; Hou, W.; Goldstein, L.A.; Lu, C.; Stolz, D.B.; Yin, X.-M.; Rabinowich, H. Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells. J. Biol. Chem. 2008, 283, 19665–19677. [Google Scholar] [CrossRef] [Green Version]
  49. Fröhlich, L.F.; Mrakovcic, M.; Smole, C.; Zatloukal, K. Molecular mechanism leading to SAHA-induced autophagy in tumor cells: evidence for a p53-dependent pathway. Cancer Cell Int. 2016, 16, 68. [Google Scholar] [CrossRef] [Green Version]
  50. Fröhlich, L.F.; Mrakovcic, M.; Smole, C.; Lahiri, P.; Zatloukal, K. Epigenetic silencing of apoptosis-inducing gene expression can be efficiently overcome by combined SAHA and TRAIL treatment in uterine sarcoma cells. PLoS ONE 2014, 9, e91558. [Google Scholar] [CrossRef]
  51. White, E. The role of autophagy in cancer. J. Clin. Investig. 2015, 125, 42–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Duprez, L.; Wirawan, E.; Vanden Berghe, T.; Vandenabeele, P. Major cell death pathways at a glance. Microbes Infect. 2009, 11, 1050–1062. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, S.; Rehman, S.K.; Zhang, W.; Wen, A.; Yao, L.; Zhang, J. Autophagy is a therapeutic target in anticancer drug resistance. Biochim. Biophys. Acta 2010, 1806, 220–229. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, Y.; Hu, L.; Zheng, H.; Mao, C.; Hu, W.; Xiong, K.; Wang, F.; Liu, C. Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol. Sin. 2013, 34, 625–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Apel, A.; Herr, I.; Schwarz, H.; Rodemann, H.; Mayer, A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008, 68, 1485–1494. [Google Scholar] [CrossRef] [Green Version]
  56. Maycotte, P.; Aryal, S.; Cummings, C.; Thorburn, J.; Morgan, M.; Thorburn, A. Chloroquine sensitizes breast cancer cells to chemotherapy independent of autophagy. Autophagy 2012, 8, 200–212. [Google Scholar] [CrossRef] [Green Version]
  57. Li, L.; Xie, W.; Pan, D.; Chen, H.; Zhang, L. Inhibition of autophagy by bafilomycin A1 promotes chemosensitivity of gastric cancer cells. Tumour Biol. 2016, 37, 653–659. [Google Scholar] [CrossRef]
  58. Carew, J.S.; Nawrocki, S.T.; Kahue, C.N.; Zhang, H.; Yang, C.; Chung, L.; Houghton, J.A.; Huang, P.; Giles, F.J.; Cleveland, J.L. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 2007, 110, 313–323. [Google Scholar] [CrossRef] [Green Version]
  59. Carew, J.S.; Kelly, K.R.; Nawrocki, S.T. Autophagy as a target for cancer therapy: new developments. Cancer Manag. Res. 2012, 4, 357–365. [Google Scholar] [CrossRef] [Green Version]
  60. Carew, J.S.; Nawrocki, S.T.; Cleveland, J.L. Modulating autophagy for therapeutic benefit. Autophagy 2007, 3, 464–467. [Google Scholar] [CrossRef] [Green Version]
  61. Amaravadi, R.; Kimmelmann, A.; Debnath, J. Targeting autophagy in cancer: recent advances and future directions. Cancer Discov. 2019, 9, 1167–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Torgersen, M.L.; Engedal, N.; Bøe, S.; Hokland, P.; Simonsen, A. Targeting autophagy potentiates the apoptotic effect of histone deacetylase inhibitors in t(8; 21) AML cells. Blood 2018, 122, 2467–2477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Galluzzi, L.; Bravo-San Pedro, J.M.; Levine, B.; Green, D.R.; Kroemer, G. Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 2017, 16, 487–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Stankov, M.V.; El Khatib, M.; Kumar Thakur, B.; Heitmann, K.; Panayotova-Dimitrova, D.; Schoening, J.; Bourquin, J.P.; Schweitzer, N.; Leverkus, M.; Welte, K.; et al. Histone deacetylase inhibitors induce apoptosis in myeloid leukemia by suppressing autophagy. Leukemia 2014, 28, 577–588. [Google Scholar] [CrossRef] [Green Version]
  65. Carew, J.S.; Medina, E.C.; Esquivel, J.A.; Mahalingam, D.; Swords, R.; Kelly, K.; Zhang, H.; Huang, P.; Mita, A.C.; Mita, M.M.; et al. Autophagy inhibition enhances vorinostat-induced apoptosis via ubiquitinated protein accumulation. J. Cell. Mol. Med. 2010, 14, 2448–2459. [Google Scholar] [CrossRef]
  66. Kumar, A.; Singh, K. Targeting autophagy to overcome drug resistance in cancer therapy. Future Med. Chem. 2015, 7, 1535–1542. [Google Scholar] [CrossRef]
  67. Gammoh, N.; Marks, P.A.; Jiang, X. Curbing autophagy and histone deacetylases to kill cancer cells. Autophagy 2012, 8, 1521–1522. [Google Scholar] [CrossRef]
  68. Ying-Jie, L.; Yu-He, L.; Yao, N.; Wang, C.-R.; Hu, N.; Ye, W.-C.; Zhang, D.-M.; Chen, Z.-S. Autophagy and multidrug resistance in cancer. Chin. J. Cancer 2017, 36, 52. [Google Scholar] [CrossRef]
  69. Li, X.; Zhou, Y.; Li, Y.; Yang, L.; Ma, Y.; Peng, X.; Yang, S. Autophagy: A novel mechanism of chemoresistance in cancers. Biomed. Pharmacother. 2019, 119, 109415. [Google Scholar] [CrossRef]
  70. Smith, A.G.; Macleod, K.F. Autophagy, cancer stem cells and drug resistance. J. Pathol. 2019, 247, 708–718. [Google Scholar] [CrossRef] [Green Version]
  71. Qadir, M.; Kwok, B.; Dragowska, W.; To, K.; Le, D.; Bally, M.; Gorski, S. Macroautophagy inhibition sensitizes tamoxifen-resistant breast cancer cells and enhances mitochondrial depolarization. Breast Cancer Res. Treat. 2008, 112, 389–403. [Google Scholar] [CrossRef] [PubMed]
  72. Samaddar, J.; Gaddy, V.; Duplantier, J.; Thandavan, S.; Shah, M.; Smith, M.; Browning, D.; Rawson, J.; Smith, S.; Barrett, J.; et al. A role for macroautophagy in protection against 4-hydroxytamoxifen-induced cell death and the development of antiestrogen resistance. Mol. Cancer Ther. 2008, 7, 2977–2987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nguyen, H.; Yang, J.; Kung, H.; Shi, X.; Tilki, D.; Lara, P.J.; DeVere Whie, R.; Gao, A.; Evans, C. Targeting autphagy overcomes Enzalutamide resistance in castration-resistant prostate cancer cells and improves therapeutic response in a xenograft model. Oncogene 2014, 33, 4521–4530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Gupta, A.; Roy, S.; Lazar, A.; Wang, W.; McAuliffe, J.; Reynoso, D.; McMahon, J.; Taguchi, T.; Floris, G.; Debiec-Rychter, M.; et al. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST). Proc. Natl. Acad. Sci. USA 2010, 107, 14333–14338. [Google Scholar] [CrossRef] [Green Version]
  75. Amaravadi, R.; Lippincott-Schwartz, J.; Yin, X.; Weiss, W.; Takebe, N.; Timmer, W.; DiPaola, R.; Lotze, M.; White, E. Principles and current strategies for targeting autophagy for cancer treatment. Clin Cancer Res. 2011, 17, 654–666. [Google Scholar] [CrossRef] [Green Version]
  76. Crighton, D.; Wilkinson, S.; O’Prey, J.; Syed, N.; Smith, P.; Harrison, P.R.; Gasco, M.; Garrone, O.; Crook, T.; Ryan, K.M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126, 121–134. [Google Scholar] [CrossRef] [Green Version]
  77. Crighton, D.; Wilkinson, S.; Ryan, K. DRAM links autophagy to p53 and programmed cell death. Autophagy 2007, 3, 72–74. [Google Scholar] [CrossRef] [Green Version]
  78. Liao, R.; Lin, Y.; Zhu, L. Molecular pathways involved in microRNA-mediated regulation of multidrug resistance. Mol. Biol. Rep. 2018, 45, 2913–2923. [Google Scholar] [CrossRef]
  79. Li, Y.; Seto, E. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harb. Perspect. Med. 2016, 6, a026831. [Google Scholar] [CrossRef] [Green Version]
  80. Chrun, E.; Modolo, F.; Ivan, F.D. Histone modifications: A review about the presence of this epigenetic phenomenon in carcinogenesis. Pathol. Pract. 2017, 213, 1329–1339. [Google Scholar] [CrossRef]
  81. Barneda-Zahonero, B.; Parra, M. Histone deacetylases and cancer. Mol. Oncol. 2012, 6, 579–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Grunstein, M. Histone acetylation in chromatin structure and transcription. Nature 1997, 389, 349–352. [Google Scholar] [CrossRef] [PubMed]
  83. Glozak, M.A.; Seto, E. Histone deacetylases and cancer. Oncogene 2007, 26, 5420–5432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ropero, S.; Esteller, M. The role of histone deacetylases (HDACs) in human cancer. Mol. Oncol. 2007, 1, 19–25. [Google Scholar] [CrossRef]
  85. Spange, S.; Wagner, T.; Heinzel, T.; Krämer, O.H. Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int. J. Biochem. Cell Biol. 2009, 41, 185–198. [Google Scholar] [CrossRef]
  86. Lee, D.Y.; Hayes, J.J.; Pruss, D.D.; Wolffe, A.P. A positive role for histone acetylation in transcription factor access to nucleosomal DNA. Cell 1993, 72, 73–84. [Google Scholar] [CrossRef]
  87. Eckschlager, T.; Plch, J.; Stiborova, M.; Hrabeta, J. Histone deacetylase inhibitors as anticancer drugs. Int. J. Mol. Sci. 2017, 18, 1414. [Google Scholar] [CrossRef]
  88. Xu, W.S.; Parmigiani, R.B.; Marks, P.A. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 2007, 26, 5541–5552. [Google Scholar] [CrossRef] [Green Version]
  89. Bose, P.; Dai, Y.; Grant, S. Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights. Pharmacol. Ther. 2014, 143, 323–336. [Google Scholar] [CrossRef] [Green Version]
  90. Vrana, J.; Decker, R.; Johnson, C.; Wang, Z.; Jarvis, W.; Richon, V.; Ehinger, M.; Fisher, P.; Grant, S. Induction of apoptosis in U937 human leukemia cells by suberoylanilide hydroxamic acid (SAHA) proceeds through pathways that are regulated by Bcl-2/Bcl-XL, c-Jun, and p21CIP1, but independent of p53. Oncogene 1999, 18, 7016–7025. [Google Scholar] [CrossRef] [Green Version]
  91. Nawrocki, S.T.; Carew, J.S.; Douglas, L.; Cleveland, J.L.; Humphreys, R.; Houghton, J.A. Histone deacetylase inhibitors enhance lexatumumab-induced apoptosis via a p21 Cip1 -dependent decrease in survivin levels. Cancer Res. 2007, 67, 6987–6995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Rahmani, M.; Yu, C.; Reese, E.; Ahmed, W.; Hirsch, K.; Dent, P.; Grant, S. Inhibition of PI-3 kinase sensitizes human leukemic cells to histone deacetylase inhibitor-mediated apoptosis through p44/42 MAP kinase inactivation and abrogation of p21 CIP1/WAF1 induction rather than AKT inhibition. Oncogene 2003, 22, 6231–6242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Burgess, A.J.; Pavey, S.; Warrener, R.; Hunter, L.K.; Piva, T.J.; Musgrove, E.A.; Saunders, N.; Parsons, P.G.; Gabrielli, B.G. Up-regulation of p21 WAF1/CIP1 by histone deacetylase inhibitors reduces their cytotoxicity. Mol. Pharmacol. 2001, 60, 828–837. [Google Scholar] [PubMed]
  94. Richon, V.; Emiliani, S.; Verdin, E.; Webb, Y.; Breslow, B.; Rifkind, R.; Marks, P.A. A class of hybrid polar inducers of transformed cell differentiation inhibits histone deacetylases. Proc. Natl. Acad. Sci. USA 1998, 95, 3003–3007. [Google Scholar] [CrossRef] [Green Version]
  95. Richon, V.M.; Sandhoff, T.W.; Rifkind, R.A.; Marks, P.A. Histone deacetylase inhibitor selectively induces p21 WAF1 expression and gene-associated histone acetylation. Proc. Natl. Acad. Sci. USA 2000, 97, 1–6. [Google Scholar] [CrossRef] [Green Version]
  96. Souleimani, A.; Asselin, C. Regulation of c-myc expression by sodium butyrate in the colon carcinoma cell line Caco-2. FEBS Lett. 1993, 326, 45–50. [Google Scholar] [CrossRef] [Green Version]
  97. Hirsch, C.L.; Smith-Windsor, E.L.; Bonham, K. Src family kinase members have a common response to histone deacetylase inhibitors in human colon cancer cells. Int. J. Cancer 2006, 118, 547–554. [Google Scholar] [CrossRef]
  98. Qiu, L.; Burgess, A.; Fairlie, D.; Leonard, H.; Parsons, P.; Gabrielli, B. Histone deacetylase inhibitors trigger a G2 checkpoint in normal cells that is defective in tumor cells. Mol. Biol. Cell 2000, 11, 2069–2083. [Google Scholar] [CrossRef] [Green Version]
  99. Ruefli, A.A.; Ausserlechner, M.J.; Bernhard, D.; Sutton, V.R.; Tainton, K.M.; Kofler, R.; Smyth, M.J.; Johnstone, R.W. The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of Bid and production of reactive oxygen species. Proc. Natl. Acad. Sci. USA 2001, 98, 10833–10838. [Google Scholar] [CrossRef] [Green Version]
  100. Lee, J.; Choy, M.L.; Ngo, L.; Foster, S.S.; Marks, P.A. Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc. Natl. Acad. Sci. USA 2010, 107, 14639–14644. [Google Scholar] [CrossRef] [Green Version]
  101. Warrener, R.; Beamish, H.; Burgess, A.; Waterhouse, N.J.; Giles, N.; Fairlie, D.; Gabrielli, B. Tumor cell-selective cytotoxicity by targeting cell cycle checkpoints. FASEB J. 2003, 17, 1550–1552. [Google Scholar] [CrossRef] [PubMed]
  102. Ungerstedt, J.S.; Sowa, Y.; Xu, W.; Shao, Y.; Dokmanovic, M.; Perez, G.; Ngo, L.; Holmgren, A.; Jiang, X.; Marks, P.A. Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc. Natl. Acad. Sci. USA 2005, 102, 673–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Butler, L.M.; Zhou, X.; Xu, W.-S.; Scher, H.I.; Rifkind, R.A.; Marks, P.A.; Richon, V.M. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl. Acad. Sci. USA 2002, 99, 11700–11705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Rosato, R.R.; Almenara, J.A.; Grant, S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21 CIP1/WAF1. Cancer Res. 2003, 63, 3637–3645. [Google Scholar] [PubMed]
  105. Bali, P.; Pranpat, M.; Bradner, J.; Balasis, M.; Fiskus, W.; Guo, F.; Rocha, K.; Kumaraswamy, S.; Boyapalle, S.; Atadja, P.; et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J. Biol. Chem. 2005, 280, 26729–26734. [Google Scholar] [CrossRef] [Green Version]
  106. Lin, K.; Wang, Y.; Chen, C.; Ho, C.; Su, W.; Jou, Y. HDAC inhibitors augmented cell migration and metastasis through induction of PKCs leading to identification of low toxicity modalities for combination cancer therapy. Clin Cancer Res. 2012, 18, 4691–4701. [Google Scholar] [CrossRef] [Green Version]
  107. Pulukuri, S.; Gorantla, B.; Rao, J. Inhibition of histone deacetylase activity promotes invasion of human cancer cells through activation of urokinase plasminogen activator. J. Biol. Chem. 2007, 282, 35594–35603. [Google Scholar] [CrossRef] [Green Version]
  108. Schoepflin, Z.R.; Shapiro, I.M.; Risbud, M.V. Class I and IIa HDACs mediate HIF-1a stability through PHD2-dependent mechanism, while HDAC6, a class IIb member, promotes HIF1a transcriptional activity in nucleus pulposus cells of the intervertebral disc. J. Bone Miner. Res. 2016, 31, 1287–1299. [Google Scholar] [CrossRef] [Green Version]
  109. Deroanne, C.F.; Bonjean, K.; Servotte, S.; Devy, L.; Colige, A.; Clausse, N.; Blacher, S.; Verdin, E.; Foidart, J.M.; Nusgens, B.V.; et al. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 2002, 21, 427–436. [Google Scholar] [CrossRef] [Green Version]
  110. Jeong, J.W.; Bae, M.K.; Ahn, M.Y.; Kim, S.H.; Sohn, T.K.; Bae, M.H.; Yoo, M.A.; Song, E.J.; Lee, K.J.; Kim, K.W. Regulation and destabilization of HIF-1alpha by ARD1-mediated acetylation. Cell 2002, 111, 709–720. [Google Scholar] [CrossRef] [Green Version]
  111. Kim, M.S.; Kwon, H.J.; Lee, Y.M.; Baek, J.H.; Jang, J.-E.; Lee, S.-W.; Moon, E.-J.; Kim, H.-S.; Lee, S.-K.; Chung, H.Y.; et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat. Med. 2001, 7, 437–443. [Google Scholar] [CrossRef]
  112. Liu, T.; Kuljaca, S.; Tee, A.; Marshall, G.M. Histone deacetylase inhibitors: multifunctional anticancer agents. Cancer Treat. Rev. 2006, 32, 157–165. [Google Scholar] [CrossRef] [PubMed]
  113. Joseph, J.; Mudduluru, G.; Antony, S.; Vashistha, S.; Ajitkumar, P.; Somasundaram, K. Expression profiling of sodium butyrate (nab)-treated cells: Identification of regulation of genes related to cytokine signaling and cancer metastasis by nab. Oncogene 2004, 23, 6304–6315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. McGarry, L.; Winnie, J.; Ozanne, B. Invasion of v-fos(fbr)-transformed cells is dependent upon histone deacetylase activity and suppression of histone deacetylase regulated genes. Oncogene 2004, 23, 5284–5392. [Google Scholar] [CrossRef] [Green Version]
  115. Kim, S.; Ahn, S.; Han, J.; Lee, H.; Lee, H.; Lee, Y.; Kim, M.; Kim, K.; Kim, W.; Hong, S. Apicidin is a histone deacetylase inhibitor with anti-invasive and anti-angiogenic potentials. Biochem. Biophys. Res. Commun. 2004, 315, 964–970. [Google Scholar] [CrossRef]
  116. Munshi, A.; Kurland, J.; Nishikawa, T.; Tanaka, T.; Hobbs, M.L.; Tucker, S.L.; Ismail, S.; Stevens, C.; Meyn, R. Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity. Clin. Cancer Res. 2005, 11, 4912–4922. [Google Scholar] [CrossRef] [Green Version]
  117. Subramanian, C.; Opipari, A.W.; Bian, X.; Castle, V.P.; Kwok, R.P. Ku70 acetylation mediates neuroblastoma cell death induced by histone deacetylase inhibitors. Proc. Natl. Acad. Sci. USA 2005, 102, 4842–4847. [Google Scholar] [CrossRef] [Green Version]
  118. Adimoolam, S.; Sirisawad, M.; Chen, J.; Thiemann, P.; Ford, J.M.; Buggy, J.J. HDAC inhibitor PCI-24781 decreases RAD51 expression and inhibits homologous recombination. Proc. Natl. Acad. Sci. USA 2007, 104, 19482–19487. [Google Scholar] [CrossRef] [Green Version]
  119. Namdar, M.; Perez, G.; Ngo, L.; Marks, P.A. Selective inhibition of histone deacetylase 6 (HDAC6) induces DNA damage and sensitizes transformed cells to anticancer agents. Proc. Natl. Acad. Sci. USA 2010, 107, 20003–20008. [Google Scholar] [CrossRef] [Green Version]
  120. Tang, Y.; Zhao, W.; Chen, Y.; Zhao, Y.; Gu, W. Acetylation Is Indispensable for p53 Activation. Cell 2008, 133, 612–626. [Google Scholar] [CrossRef] [Green Version]
  121. Mrakovcic, M.; Fröhlich, L.F. p53-mediated molecular control of autophagy in tumor cells. Biomolecules 2018, 8, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Mrakovcic, M.; Bohner, L.; Hanisch, M.; Fröhlich, L.F. Epigenetic targeting of autophagy via HDAC inhibition in tumor cells: Role of p53. Int. J. Mol. Sci. 2018, 19, 3952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Mrakovcic, M.; Kleinheinz, J.; Fröhlich, L.F. Histone deacetylase inhibitor-induced autophagy in tumor cells: Implications for p53. Int. J. Mol. Sci. 2017, 18, 1883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mrakovcic, M.; Fröhlich, L. Regulation of HDACi-triggered autophagy by the tumor suppressor protein p53. In Genes and Cancer; Lemamy, G.-J., Ed.; InTech Open: Rijeka, Croatia, 2019; pp. 1–26. ISBN 978-1-78984-427-6. [Google Scholar]
  125. Koeneke, E.; Witt, O.; Oehme, I. HDAC family members intertwined in the regulation of autophagy: A druggable vulnerability in aggressive tumor entities. Cells 2015, 4, 135–168. [Google Scholar] [CrossRef] [PubMed]
  126. Carew, J.S.; Giles, F.J.; Nawrocki, S.T. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett. 2008, 269, 7–17. [Google Scholar] [CrossRef]
  127. Rikiishi, H. Autophagic and apoptotic effects of HDAC inhibitors on cancer cells. J. Biomed. Biotechnol. 2011, 2011, 830260. [Google Scholar] [CrossRef]
  128. Di Giacomo, V.; Di Valerio, V.; Rapino, M.; Bosco, D.; Cacciatore, I.; Ciulla, M.; Marrazzo, A.; Fiorito, J.; Di Stefano, A.; Cataldi, A. MRJF4, a novel histone deacetylase inhibitor, induces p21 mediated autophagy in PC3 prostate cancer cells. Cell. Mol. Biol. 2015, 61, 17–23. [Google Scholar]
  129. Yamamoto, S.; Tanaka, K.; Sakimura, R.; Okada, T.; Nakamura, T.; Li, Y.; Takasaki, M.; Nakabeppu, Y.; Iwamoto, Y. Suberoylanilide hydroxamic acid (SAHA) induces apoptosis or autophagy-associated cell death in chondrosarcoma cell lines. Anticancer Res. 2008, 28, 1585–1591. [Google Scholar]
  130. Saveria, M.; Montani, G.; Granato, M.; Santoni, C.; Del Porto, P.; Merendino, N.; Orazi, G.D.; Faggioni, A.; Cirone, M. Histone deacetylase inhibitors VPA and TSA induce apoptosis and autophagy in pancreatic cancer cells. Cell Oncol. 2017, 40, 167–180. [Google Scholar] [CrossRef]
  131. Wang, J.; Kim, T.H.; Ahn, M.Y.; Lee, J.; Jung, J.H.; Choi, W.S.; Lee, B.M.; Yoon, K.S.; Yoon, S.; Kim, H.S. Sirtinol, a class III HDAC inhibitor, induces apoptotic and autophagic cell death in MCF-7 human breast cancer cells. Int. J. Oncol. 2012, 41, 1101–1109. [Google Scholar] [CrossRef] [Green Version]
  132. Park, E.Y.; Woo, Y.; Kim, S.J.; Kim, D.H.; Lee, E.K.; De, U.; Kim, K.S.; Lee, J.; Jung, J.H.; Ha, K.T.; et al. Anticancer effects of a new SIRT inhibitor, MHY2256, against human breast cancer MCF-7 cells via regulation of MDM2-p53 binding. Int. J. Biol. Sci. 2016, 12, 1555–1567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. De, U.; Son, J.Y.; Sachan, R.; Park, Y.J.; Kang, D.; Yoon, K.; Lee, B.M.; Kim, I.S.; Moon, H.R.; Kim, H.S. A new synthetic histone deacetylase inhibitor, MHY2256, induces apoptosis and autophagy cell death in endometrial cancer cells via p53 acetylation. Int. J. Mol. Sci. 2018, 19, 2743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ahn, M.Y.; Ahn, J.W.; Kim, H.S.; Lee, J.; Yoon, J.H. Apicidin inhibits cell growth by downregulating IGF-1R in salivary mucoepidermoid carcinoma cells. Oncol. Rep. 2015, 33, 1899–1907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Shao, Y.; Gao, Z.; Marks, P.A.; Jiang, X. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc. Natl. Acad. Sci. USA 2004, 101, 18030–18035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Gammoh, N.; Lam, D.; Puente, C.; Ganley, I.; Marks, P.A.; Jiang, X. Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc. Natl. Acad. Sci. USA 2012, 109, 6561–6565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Li, J.; Liu, R.; Lei, Y.; Wang, K.; Lau, Q.C.; Xie, N.; Zhou, S.; Nie, C.; Chen, L.; Wei, Y.; et al. Proteomic analysis revealed association of aberrant ROS signaling with suberoylanilide hydroxamic acid-induced autophagy in Jurkat T-leukemia cells. Autophagy 2010, 6, 711–724. [Google Scholar] [CrossRef] [Green Version]
  138. Chiao, M.-T.; Cheng, W.-Y.; Yang, Y.-C.; Shen, C.-C.; Ko, J.-L. Suberoylanilide hydroxamic acid (SAHA) causes tumor growth slowdown and triggers autophagy in glioblastoma stem cells. Autophagy 2013, 9, 1509–1526. [Google Scholar] [CrossRef] [Green Version]
  139. Liu, Y.L.; Yang, P.; Shun, C.; Wu, M.; Weng, J.; Chen, C. Autophagy potentiates the anti-cancer effects of the histone deacetylase inhibitors in hepatocellular carcinoma. Autophagy 2010, 6, 1057–1065. [Google Scholar] [CrossRef] [Green Version]
  140. Hrzenjak, A.; Moinfar, F.; Kremser, M.-L.; Strohmeier, B.; Petru, E.; Zatloukal, K.; Denk, H. Histone deacetylase inhibitor vorinostat suppresses the growth of uterine sarcomas in vitro and in vivo. Mol. Cancer 2010, 9, 49. [Google Scholar] [CrossRef] [Green Version]
  141. Shulak, L.; Beljanski, V.; Chiang, C.; Dutta, S.M.; Van Grevenynghe, J.; Belgnaoui, S.M.; Nguyen, T.L.A.; Di Lenardo, T.; Semmes, O.J.; Lin, R.T.; et al. Histone deacetylase inhibitors potentiate vesicular stomatitis virus oncolysis in prostate cancer cells by modulating NF-kappa B-dependent autophagy. J. Virol. 2014, 88, 2927–2940. [Google Scholar] [CrossRef] [Green Version]
  142. Long, J.; Zhao, J.; Yan, Z.; Liu, Z.; Wang, N. Antitumor effects of a novel sulfur-containing hydroxamate histone deacetylase inhibitor H40. Int. J. Cancer 2009, 124, 1235–1244. [Google Scholar] [CrossRef] [PubMed]
  143. Watanabe, M.; Adachi, S.; Matsubara, H.; Imai, T.; Yui, Y.; Mizushima, Y. Induction of autophagy in malignant rhabdoid tumor cells by the histone deacetylase inhibitor FK228 through AIF translocation. Int. J. Cancer Res. 2009, 67, 55–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ellis, L.; Bots, M.; Lindemann, R.K.; Bolden, J.E.; Newbold, A.; Cluse, L.A.; Scott, C.L.; Strasser, A.; Atadja, P.; Lowe, S.W.; et al. The histone deacetylase inhibitors LAQ824 and LBH589 do not require death receptor signaling or a functional apoptosome to mediate tumor cell death or therapeutic efficacy. Blood 2009, 114, 380–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zhang, J.; Ng, S.; Wang, J.; Zhou, J.; Tan, S.; Yang, N.; Lin, Q.; Xia, D.; Shen, H. Histone deacetylase inhibitors induce autophagy through FOXO1-dependent pathways. Autophagy 2015, 11, 629–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Sun, W.; Yi, Y.; Xia, G.; Zhao, Y.; Yu, Y.; Li, L.; Hua, C.; He, B.; Yang, B.; Yu, C.; et al. Nrf2-miR-129-3p-mTOR Axis Controls an miRNA Regulatory Network Involved in HDACi-Induced Autophagy. Mol. Ther. 2019, 27, 1039–1050. [Google Scholar] [CrossRef] [PubMed]
  147. Gandesiri, M.; Chakilam, S.; Ivanovska, J.; Benderska, N.; Ocker, M.; Di Fazio, P.; Feoktistova, M.; Gali-Muhtasib, H.; Rave-Fränk, M.; Prante, O.; et al. DAPK plays an important role in panobinostat-induced autophagy and commits cells to apoptosis under autophagy deficient conditions. Apoptosis 2012, 17, 1300–1315. [Google Scholar] [CrossRef]
  148. Yi, C.; Ma, M.; Ran, L.; Zheng, J.; Tong, J.; Zhu, J.; Ma, C.; Sun, Y.; Zhang, S.; Feng, W.; et al. Function and molecular mechanism of acetylation in autophagy regulation. Science 2012, 336, 474–477. [Google Scholar] [CrossRef]
  149. Lee, I.H.; Cao, L.; Mostoslavsky, R.; Lombard, D.B.; Liu, J.; Bruns, N.E.; Tsokos, M.; Alt, F.W.; Finkel, T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc. Natl. Acad. Sci. USA 2008, 105, 3374–3379. [Google Scholar] [CrossRef] [Green Version]
  150. Glozak, M.A.; Sengupta, N.; Zhang, X.; Seto, E. Acetylation and deacetylation of non-histone proteins. Gene 2005, 363, 15–23. [Google Scholar] [CrossRef]
  151. El-Khoury, V.; Pierson, S.; Szwarcbart, E.; Brons, N.H.C.; Roland, O.; Cherrier-De Wilde, S.; Plawny, L.; Van Dyck, E.; Berchem, G. Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia 2014, 28, 1636–1646. [Google Scholar] [CrossRef] [Green Version]
  152. El-Khoury, V.; Moussay, E.; Janji, B.; Palissot, V.; Aouali, N.; Brons, N.H.; Van Moer, K.; Pierson, S.; Van Dyck, E.; Berchem, G. The histone deacetylase inhibitor MGCD0103 induces apoptosis in B-cell chronic lymphocytic leukemia cells through a mitochondria-mediated caspase activation cascade. Mol. Cancer Ther. 2010, 9, 1349–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Hrzenjak, A.; Kremser, M.; Strohmeier, B.; Moinfar, F.; Zatloukal, K.; Denk, H. SAHA induces caspase-independent, autophagic cell death of endometrial stromal sarcoma cells by influencing the mTOR pathway. J. Pathol. 2008, 216, 495–504. [Google Scholar] [CrossRef] [PubMed]
  154. Cao, Q.; Yu, C.; Xue, R.; Hsueh, W.; Pan, P.; Chen, Z.; Wang, S.; McNutt, M.; Gu, J. Autophagy induced by suberoylanilide hydroxamic acid in Hela S3 cells involves inhibition of protein kinase B and up-regulation of Beclin 1. Int. J. Biochem. Cell Biol. 2008, 40, 272–283. [Google Scholar] [CrossRef] [PubMed]
  155. Podsypanina, K.; Lee, R.; Politis, C.; Hennessey, I.; Crance, A.; Puc, J.; Neshat, M.; Wang, H.; Yang, L.; Gibbons, J.; et al. An inhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinase activity in Pten+/- mice. Proc. Natl. Acad. Sci. USA 2001, 98, 10320–10325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Neshat, M.; Mellinghoff, I.; Tran, C.; Stiles, B.; Thomas, G.; Petersen, R.; Frost, P.; Gibbons, J.; Wu, H.; Sawyers, C. Enhanced sensitivity of PTEN-deficient tumors to inhibition of FRAP/mTOR. Proc. Natl. Acad. Sci. USA 2001, 98, 10314–10319. [Google Scholar] [CrossRef] [Green Version]
  157. Arico, S.; Petiot, A.; Bauvy, C.; Dubbelhuis, P.F.; Meijer, A.J.; Codogno, P.; Ogier-Denis, E. The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J. Biol. Chem. 2001, 276, 35243–35246. [Google Scholar] [CrossRef] [Green Version]
  158. Lu, Z.; Luo, R.Z.; Lu, Y.; Zhang, X.; Yu, Q.; Khare, S.; Kondo, S.; Kondo, Y.; Yu, Y.; Mills, G.B.; et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J. Clin. Investig. 2008, 118, 3917–3929. [Google Scholar] [CrossRef] [Green Version]
  159. Ahn, M.-Y.; Ahn, S.-G.; Yoon, J.-H. Apicidin, a histone deaceylase inhibitor, induces both apoptosis and autophagy in human oral squamous carcinoma cells. Oral Oncol. 2011, 47, 1032–1038. [Google Scholar] [CrossRef]
  160. Liang, X.H.; Jackson, S.; Seaman, M.; Brown, K.; Kempkes, B.; Hibshoosh, H.; Levine, B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999, 402, 672–676. [Google Scholar] [CrossRef]
  161. Reed, J.C.; Cuddy, M.; Slabiak, T.; Croce, C.M.; Nowell, P.C. Oncogenic potential of bcl-2 demonstrated by gene transfer. Nature 1988, 336, 259–261. [Google Scholar] [CrossRef]
  162. Reed, J.C.; Kitada, S.; Takayama, S.; Miyashita, T. Regulation of chemoresistance by the bcl-2 oncoprotein in non-Hodgkin’s lymphoma and lymphocytic leukemia cell lines. Ann. Oncol. 1994, 5, 61–65. [Google Scholar] [CrossRef] [PubMed]
  163. Kitada, S.; Takayama, S.; De Riel, K.; Tanaka, S.; Reed, J.C. Reversal of chemoresistance of lymphoma cells by antisense-mediated reduction of bcl-2 gene expression. Antisense Res. Dev. 1994, 4, 71–79. [Google Scholar] [CrossRef] [PubMed]
  164. Cui, Q.; Wang, J.-Q.; Assaraf, Y.G.; Ren, L.; Gupta, P.; Wei, L.; Ashby, C.R.J.; Yang, D.-H.; Chen, Z.-S. Modulating ROS to overcome multidrug resistance in cancer. Drug Resist. Updat. 2018, 41, 1–25. [Google Scholar] [CrossRef] [PubMed]
  165. Hui, K.F.; Yeung, P.L.; Chiang, A.K.S. Induction of MAPK- and ROS-dependent autophagy and apoptosis in gastric carcinoma by combination of romidepsin and bortezomib. Oncotarget 2015, 7, 1–14. [Google Scholar] [CrossRef] [Green Version]
  166. Zhan, Y.; Gong, K.; Chen, C.; Wang, H.; Li, W. P38 MAP kinase functions as a switch in MS-275-induced reactive oxygen species-dependent autophagy and apoptosis in human colon cancer cells. Free Radic. Biol. Med. 2012, 53, 532–543. [Google Scholar] [CrossRef]
  167. Dittmer, D.; Pati, S.; Zambetti, G.; Chu, S.; Teresky, A.K.; Moore, M.; Finlay, C.; Levine, A.J. Gain of function mutations in p53. Nat. Genet. 1993, 4, 42–46. [Google Scholar] [CrossRef]
  168. Santoro, R.; Strano, S.; Blandino, G. Transcriptional regulation by mutant p53 and oncogenesis. Subcell. Biochem. 2014, 85, 91–103. [Google Scholar] [CrossRef]
  169. Willis, A.; Jung, E.J.; Wakefield, T.; Chen, X. Mutant p53 exerts a dominant negative effect by preventing wild-type from binding to the promoter of its target genes. Oncogene 2004, 23, 2330–2338. [Google Scholar] [CrossRef] [Green Version]
  170. Zheng, T.; Wang, J.; Zhao, Y.; Zhang, C.; Lin, M.; Wang, X.; Yu, H.; Liu, L.; Feng, Z.; Hu, W. Spliced MDM2 isoforms promote mutant p53 accumulation and gain-of-function in tumorigenesis. Nat. Commun. 2013, 4, 2996. [Google Scholar] [CrossRef]
  171. Yue, X.; Zhao, Y.; Xu, Y.; Zheng, M. Mutant p53 in cancer: Accumulation, gain-of-function, and therapy. J. Mol. Biol. 2017, 429, 1595–1606. [Google Scholar] [CrossRef]
  172. Abbas, T.; Dutta, A. p21 in cancer: intricate networks and multiple activities. Nat. Rev. Cancer 2009, 9, 400–414. [Google Scholar] [CrossRef] [PubMed]
  173. Blagosklonny, M. Are p27 and p21 cytoplasmic oncoproteins? Cell Cycle 2002, 1, 391–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Abukhdeir, A.; Park, B. P21 and p27: roles in carcinogenesis and drug resistance. Expert Rev. Mol. Med. 2008, 10, e19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Xue, L.; Zhou, B.; Liu, X.; Heung, Y.; Chau, J.; Chu, E.; Li, S.; Jiang, C.; Un, F.; Yen, Y. Ribonucleotide reductase small subunit p53R2 facilitates p21 induction of G1 arrest under UV irradiation. Cancer Res. 2007, 67, 16–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Compagno, M.; Lim, W.; Grunn, A.; Nandula, S.; Brahmachary, M.; Shen, Q.; Bertoni, F.; Ponzoni, M.; Scandurra, M.; Califano, A.; et al. Mutations of multiple genes cause deregulation of NF-kB in diffuse large B cell lymphoma. Nature 2009, 459, 717–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Han, C.; Cho, K.; Choi, H.; Han, H.; Kang, K. Role of FoxO1 activation in MDR1 expression in adriamycin-resistant breast cancer cells. Carcinogenesis 2008, 29, 1837–1844. [Google Scholar] [CrossRef] [Green Version]
  178. Goto, T.; Takano, M.; Hirata, J.; Tsuda, H. The involvement of FOXO1 in cytotoxic stress and drug-resistance induced by paclitaxel in ovarian cancers. Br. J. Cancer 2008, 98, 1068–1075. [Google Scholar] [CrossRef]
  179. Ramanathan, B.; Jan, K.; Chen, C.; Hour, T.; Yu, H.; Pu, Y. Resistance to paclitaxel is proportional to cellular total antioxidant capacity. Cancer Res. 2005, 65, 8455–8460. [Google Scholar] [CrossRef] [Green Version]
  180. Ogawa, T.; Liggett, T.; Melnikov, A.; Monitto, C.; Kusuke, D.; Shiga, K.; Kobayashi, T.; Horii, A.; Chatterjee, A.; Levenson, V.; et al. Methylation of death-associated protein kinase is associated with cetuximab and erlotinib resistance. Cell Cycle 2012, 11, 1656–1663. [Google Scholar] [CrossRef] [Green Version]
  181. Lee, Y.-R.; Yuan, W.-C.; Ho, H.-C.; Chen, C.-H.; Shih, H.-M.; Chen, R.-H. The Cullin 3 substrate adaptor KLHL20 mediates DAPK ubiquitination to control interferon responses. EMBO J. 2010, 29, 1748–1761. [Google Scholar] [CrossRef]
  182. Yamamoto, M.; Kensler, T.; Motohashi, H. The KEAP1-NRF2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiol. Rev. 2018, 98, 1169–1203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Ann. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. DeNicola, G.M.; Karreth, F.A.; Humpton, T.J.; Gopinathan, A.; Wei, C.; Frese, K.; Mangal, D.; Yu, K.H.; Yeo, C.J.; Calhoun, E.S.; et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 2011, 475, 106–109. [Google Scholar] [CrossRef] [PubMed]
  185. Robey, R.W.; Chakraborty, A.R.; Basseville, A.; Luchenko, V.; Zhan, Z.; Bates, S.E. Histone deacetylase inhibitors: emerging mechanisms of resistance. Mol. Pharmacol. 2012, 8, 2021–2031. [Google Scholar] [CrossRef]
  186. Fantin, V.; Richon, V. Mechanisms of resistance to histone deacetylase inhibitors and their therapeutic implications. Clin Cancer Res. 2007, 13, 7237–7242. [Google Scholar] [CrossRef] [Green Version]
  187. Lopez, G.; Torres, K.; Liu, J.; Hernandez, B.; Young, E.; Belousov, R.; Bolshakov, S.; Lazar, A.J.; Slopis, J.M.; McCutcheon, I.E.; et al. Autophagic survival in resistance to histone deacetylase inhibitors: novel strategies to treat malignant peripheral nerve sheath tumors. Cancer Res. 2011, 71, 185–196. [Google Scholar] [CrossRef] [Green Version]
  188. Lee, J.-H.; Choy, M.L.; Marks, P.A. Mechanism of resistance to histone deacetylase inhibitors. Adv. Cancer Res. 2012, 116, 39–86. [Google Scholar] [CrossRef]
  189. Hippert, M.M.; Toole, P.S.O.; Thorburn, A. Autophagy in cancer: Good, bad, or both? Cancer Res. 2006, 66, 9349–9352. [Google Scholar] [CrossRef] [Green Version]
  190. Torgersen, M.L.; Simonsen, A. Autophagy-Friend or foe in the treatment of fusion protein-associated leukemias? Autophagy 2013, 9, 2175–2177. [Google Scholar] [CrossRef] [Green Version]
  191. Robey, R.; Zhan, Z.; Piekarz, R.; Kayastha, G.; Fojo, T.; Bates, S. Increased MDR1 expression in normal and malignant peripheral blood mononuclear cells obtained from patients receiving depsipeptide (FR901228, FK228, NSC630176). Clin. Cancer Res. 2006, 12, 1547–1555. [Google Scholar] [CrossRef] [Green Version]
  192. Piekarz, R.; Robey, R.; Zhan, Z.; Kayastha, G.; Sayah, A.; Abdeldaim, A.; Torrico, S.; Bates, S. T-cell lymphoma as a model for the use of histone deacetylase inhibitors in cancer therapy: impact of depsipeptide on molecular markers, therapeutic targets, and mechanisms of resistance. Blood 2004, 103, 4636–4643. [Google Scholar] [CrossRef] [PubMed]
  193. Xiao, J.; Huang, Y.; Dai, Z.; Sadee, W.; Chen, J.; Liu, S.; Marcucci, G.; Byrd, J.; Covey, J.; Wright, J.; et al. Chemoresistance to depsipeptide FK228 is mediated by reversible MDR1 induction in human cancer cell lines. J. Pharmacol. Exp. Ther. 2005, 314, 467–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Epping, M.; Meijer, L.; Bos, J.; Bernards, R. UNC45A confers resistance to histone deacetylase inhibitors and retinoic acid. Mol. Cancer Res. 2009, 7, 1861–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Mickley, L.; Bates, S.; Richert, N.; Currier, S.; Tanaka, S.; Foss, F.; Rosen, N.; Fojo, A. Modulation of the expression of a multidrug resistance gene (mdr-1/P-gylcoprotein) by differentiating agents. J. Biol. Chem. 1989, 264, 18301–18340. [Google Scholar]
  196. Frommel, T.; Coon, J.; Tsuruo, T.; Roninson, I. Variable effects of sodium butyrate on the expression and function of the mdr-1 (P-glycoprotein) gene in colon carcinoma cell lines. Int. J. Cancer 1993, 55, 297–302. [Google Scholar] [CrossRef]
  197. Fantin, V.; Loboda, A.; Paweletz, C.; Hendrickson, R.; Pierce, J.; Roth, J.; Li, L.; Gooden, F.; Korenchuck, S.; Hou, X.; et al. Constitutive activation of signal transducers and activators of transcription predicts vorinostat resistance in cutaneous T-cell lymphoma. Cancer Res. 2008, 68, 3785–3794. [Google Scholar] [CrossRef] [Green Version]
  198. Paquette, M.; El-Houjeiri, L.; Pause, A. mTOR pathways in cancer and autophagy. Cancers 2018, 10, 18. [Google Scholar] [CrossRef] [Green Version]
  199. Noda, T. Regulation of Autophagy through TORC1 and mTORC1. Biomolecules 2017, 7, 52. [Google Scholar] [CrossRef]
  200. Yin, Y.; Hua, H.; Li, M.; Liu, S.; Kong, Q.; Shao, T.; Wang, J.; Luo, Y.; Wang, Q.; Luo, T.; et al. mTORC2 promotes type I insulin-like growth factor receptor and insulin receptor activation through the tyrosine kinase activity of mTOR. Cell Res. 2016, 26, 46–65. [Google Scholar] [CrossRef] [Green Version]
  201. Saxton, R.; Sabatini, D. mTOR signaling in growth, metabolism, and disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [Green Version]
  202. Laplante, M.; Sabatini, D. mTOR signaling. Cold Spring Harb. Perspect. Biol. 2012, 4, a011593. [Google Scholar] [CrossRef] [PubMed]
  203. Loewith, R.; Jacinto, E.; Wullschlager, S.; Lorberg, A.; Crespo, J.; Bonenfant, D.; Oppliger, W.; Jenoe, P.; Hall, M. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 2002, 10, 457–468. [Google Scholar] [CrossRef]
  204. Ma, X.; Blenis, J. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell Biol. 2009, 10, 307–318. [Google Scholar] [CrossRef] [PubMed]
  205. Sun, S.Y.; Rosenberg, L.M.; Wang, X.; Zhou, Z.; Yue, P.; Fu, H.; Khuri, F.R. Activation of Akt and elF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. Cancer Res. 2005, 65, 7052–7058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Shackelford, D.B.; Shaw, R.J. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat. Rev. Cancer 2009, 9, 563–575. [Google Scholar] [CrossRef]
  207. Luo, Z.; Saha, A.K.; Xiang, X.; Ruderman, N.B. AMPK, the metabolic syndrome and cancer. Trends Pharmacol. Sci. 2005, 26, 69–76. [Google Scholar] [CrossRef]
  208. Kim, J.; Kundu, M.; Viollet, B.; Kun-Liang, G. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [Green Version]
  209. Jung, C.H.; Jun, C.B.; Ro, S.; Kim, Y.; Otto, N.M.; Cao, J.; Kundu, M.; Kim, D. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol. Biol. Cell 2009, 20, 1992–2003. [Google Scholar] [CrossRef] [Green Version]
  210. Shaw, R.J.; Kosmatka, M.; Bardeesy, N.; Hurley, R.L.; Witters, L.A.; DePinho, R.A.; Cantley, L.C. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc. Natl. Acad. Sci. USA 2004, 101, 3329–3335. [Google Scholar] [CrossRef] [Green Version]
  211. Lin, S.; Li, T.; Liu, Q.; Zhang, C.; Li, X.; Chen, Y.; Zhang, S.; Lian, G.; Liu, Q.; Ruan, K.; et al. GSK3-TIP60-ULK1 signaling links growth factor deprivation to autophagy. Science 2012, 336, 477–481. [Google Scholar] [CrossRef]
  212. Aoki, M.; Blazek, E.; Vogt, P. A role for the kinase mTOR in cellular transformation induced by the oncoproteins P3k and Akt. Proc. Natl. Acad. Sci. USA 2001, 98, 136–141. [Google Scholar] [CrossRef] [PubMed]
  213. Klionsky, D.J.; Meijer, A.J.; Codogno, P. Autophagy and p70S6 kinase. Autophagy 2005, 1, 59–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Balakumaran, B.S.; Herbert, J.T.; Febbo, P.G. MYC activity mitigates response to rapamycin in prostate cancer through 4EBP1-mediated inhibition of autophagy. Autophagy 2010, 6, 281–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. He, C.; Levine, B. The Beclin 1 interactome. Curr. Opin. Cell Biol. 2010, 22, 140–149. [Google Scholar] [CrossRef]
  216. Maiuri, M.; Le Toumelin, G.; Criollo, A.; Rain, J.; Gautier, F.; Juin, P.; Tasdemir, E.; Pierron, G.; Troulinaki, K.; Tavernarakis, N.; et al. Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J. 2007, 26, 2527–2539. [Google Scholar] [CrossRef]
  217. Zalckvar, E.; Berissi, H.; Mizrachy, L.; Idelchuk, Y.; Koren, I.; Eisenstein, M.; Sabanay, H.; Pinkas-Kramarski, R.; Kimchi, A. DAP-kinase mediated phosphorylation on the BH3 domain of beclin 1 promotes dissociation of beclin 1 from Bcl-XL and induction of autophagy. EMBO Rep. 2009, 10, 285–292. [Google Scholar] [CrossRef] [Green Version]
  218. Pattingre, S.; Tassa, A.; Qu, X.; Garuti, R.; Liang, X.; Mizushima, N.; Packer, M.; Schneider, M.; Levine, B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005, 122, 927–939. [Google Scholar] [CrossRef] [Green Version]
  219. Erlich, S.; Mizrachy, L.; Segev, O.; Lindenboim, L.; Zmira, O.; Adi-Harel, S.; Hirsch, J.A.; Stein, R.; Pinkas-Kramarski, R. Differential interactions between Beclin 1 and Bcl-2 family members. Autophagy 2007, 3, 561–568. [Google Scholar] [CrossRef] [Green Version]
  220. Yuan, T.; Cantley, L. PI3K pathway alterations in cancer: variations on a theme. Oncogene 2008, 27, 5497–5510. [Google Scholar] [CrossRef] [Green Version]
  221. Shaw, R.; Cantley, L. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 2006, 441, 424–430. [Google Scholar] [CrossRef]
  222. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumors. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Hua, H.; Kong, Q.; Zhang, H.; Wang, J.; Luo, T.; Jiang, Y. Targeting mTOR for cancer therapy. J. Hematol. Oncol. 2019, 12, 71. [Google Scholar] [CrossRef] [PubMed]
  224. Akar, U.; Chaves-Reyez, A.; Barria, M.; Tari, A.; Sanguino, A.; Kondo, Y.; Kondo, S.; Arun, B.; Lopez-Berestein, G.; Ozpolat, B. Silencing of Bcl-2 expression by small interfering RNA induces autophagic cell death in MCF-7 breast cancer cells. Autophagy 2008, 4, 669–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Szumiel, I. Autophagy, reactive oxygen species and the fate of mammalian cells. Free Radic. Res. 2011, 45, 253–265. [Google Scholar] [CrossRef]
  226. Zhang, J.; Ney, P. Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ. 2009, 16, 939–946. [Google Scholar] [CrossRef] [Green Version]
  227. Novak, I. Mitophagy: A complex mechanism of mitochondrial removal. Antioxidants Redox Signal. 2012, 17, 794–802. [Google Scholar] [CrossRef]
  228. Feng, D.; Liu, L.; Zhu, Y.; Chen, Q. Molecular signaling toward mitophagy and its physiological significance. Exp. Cell Res. 2013, 319, 1697–1705. [Google Scholar] [CrossRef]
  229. Ranganathan, A.; Zhang, L.; Adam, A.; Aquirre-Ghiso, J. Functional coupling of p38-induced upregulation of BiP and activation of RNA-dependent protein kinase-like endoplasmatic reticulum kinase to drug resistance of dormant carcinoma cells. Cancer Res. 2006, 66, 1702–1711. [Google Scholar] [CrossRef] [Green Version]
  230. Warr, M.; Binnewies, M.; Flach, J.; Reynaud, D.; Garg, T.; Malhotra, R.; Debnath, J.; Passegue, E. FOXO3 directs a protective autophagy program in hematopoietic stem cells. Nature 2013, 494, 323–327. [Google Scholar] [CrossRef] [Green Version]
  231. Rouschop, K.; van den Beucken, T.; Dubois, L.; Niessen, H.; Bussink, J.; Savelkouls, K.; Keulers, T.; Mujcic, H.; Landuyt, W.; Voncken, J.; et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J. Clin. Investig. 2010, 120, 127–141. [Google Scholar] [CrossRef]
  232. Alexander, A.; Cai, S.; Kim, J.; Nanez, A.; Sahin, M.; MacLean, K.; Inoki, K.; Guan, K.; Shen, J.; Person, M.; et al. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc. Natl. Acad. Sci. USA 2010, 107, 4153–4158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Scherz-Shouval, R.; Elazar, Z. Regulation of autophagy by ROS: physiology and pathology. Trends Biochem. Sci. 2011, 36, 30–38. [Google Scholar] [CrossRef] [PubMed]
  234. Takamura, A.; Komatsu, M.; Hara, T.; Sakamoto, A.; Kishi, C.; Waguri, S.; Eishi, Y.; Hino, O.; Tanaka, K.; Mizushima, N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011, 25, 795–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Mathew, R.; Kongara, S.; Beaudoin, B.; Karp, C.M.; Bray, K.; Degenhardt, K.; Chen, G.; Jin, S.; White, E. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev. 2007, 21, 1367–1381. [Google Scholar] [CrossRef] [Green Version]
  236. Karantza-Wadsworth, V.; Patel, S.; Kravchuck, O.; Chen, G.; Mathew, R.; Jin, S.; White, E. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 2007, 21, 1621–1635. [Google Scholar] [CrossRef] [Green Version]
  237. Mrakovcic, M.; Kleinheinz, J.; Fröhlich, L.F. p53 at the crossroads between different types of HDAC-inhibitor-mediated cancer cell death. Int. J. Mol. Sci. 2019, 20, 2415. [Google Scholar] [CrossRef] [Green Version]
  238. Ocker, M.; Schneider-Stock, R. Histone deacetylase inhibitors: Signalling towards p21cip1/waf1. Int. J. Biochem. Cell Biol. 2007, 39, 1367–1374. [Google Scholar] [CrossRef]
  239. Sandor, V.; Senderowicz, A.; Mertins, S.; Sackett, D.; Sausville, E.; Blagoskonny, M.V.; Bates, S.E. P21-dependent G1 arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br. J. Cancer 2000, 83, 817–825. [Google Scholar] [CrossRef]
  240. Gu, W.; Roeder, R.G. Activation of p53 Sequence-Specific DNA Binding by Acetylation of the p53 C-Terminal Domain. Cell 1997, 90, 595–606. [Google Scholar] [CrossRef] [Green Version]
  241. Wang, X.; Zhenchuk, A.; Wimann, K.; Albertioni, F. Regulation of p53R2 and its role as potential target for cancer therapy. Cancer Lett. 2009, 276, 1–7. [Google Scholar] [CrossRef]
  242. Schepers, H.; Geugien, M.; Eggen, B.; Vellenga, E. Constitutive cytoplasmic localization of p21Waf1/Cip1 affects the apoptotic process in monocytic leukaemia. Leukemia 2003, 17, 2113–2121. [Google Scholar] [CrossRef] [Green Version]
  243. Crighton, D.; O’Prey, J.; Bell, H.; Ryan, K. p73 regulates DRAM-independent autophagy that does not contribute to programmed cell death. Cell Death Differ. 2007, 14, 1071–1079. [Google Scholar] [CrossRef] [Green Version]
  244. Feng, Z. p53 regulation of the IGF-1/AKT/mTOR pathways and the endosomal compartment. Cold Spring Harb. Perspect. Biol. 2010, 2, 1–10. [Google Scholar] [CrossRef] [Green Version]
  245. Zhao, Y.; Yang, J.; Liao, W.; Liu, X.; Zhang, H.; Wang, S.; Wang, D.; Feng, J.; Yu, L.; Zhu, W.-G. Cytosolic FoxO1 is essential for the induction of autophagy and tumour suppressor activity. Nat. Cell Biol. 2010, 12, 665–675. [Google Scholar] [CrossRef]
  246. Lee, J.H.; Budanov, A.V.; Park, E.J.; Birse, R.; Kim, T.E.; Guy, A.; Ocorr, K.; Ellisman, M.H.; Bodmer, R.; Bier, E. Sestrin as a feedback inhibitor of TOR that prevents age-related pathologies. Science 2011, 327, 1223–1228. [Google Scholar] [CrossRef] [Green Version]
  247. Wang, E.Y.; Gang, H.; Aviv, Y.; Dhingra, R.; Margulets, V.; Kirshenbaum, L.A. p53 mediates autophagy and cell death by a mechanism contingent on Bnip3. Hypertension 2013, 62, 70–77. [Google Scholar] [CrossRef] [Green Version]
  248. Yee, K.S.; Wilkinson, S.; James, J.; Ryan, K.M.; Vousden, K.H. PUMA and Bax-induced autophagy contributes to apoptosis. Cell Death Differ. 2010, 16, 1135–1145. [Google Scholar] [CrossRef] [Green Version]
  249. Pimkina, J.; Humbey, O.; Zilfou, J.; Jarnik, M.; Murphy, M. ARF induces autophagy by virtue of interaction with Bcl-xl. J. Biol. Chem. 2009, 284, 2803–2810. [Google Scholar] [CrossRef] [Green Version]
  250. Balaburski, G.M.; Hontz, R.D.; Murphy, M.E. P53 and ARF: Unexpected players in autophagy. Trends Cell Biol. 2010, 20, 363–369. [Google Scholar] [CrossRef] [Green Version]
  251. Zalckvar, E.; Berissi, H.; Eisenstein, M.; Kimchi, A. Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL. Autophagy 2009, 5, 720–722. [Google Scholar] [CrossRef] [Green Version]
  252. Harrison, B.; Kraus, M.; Burch, L.; Stevens, C.; Craig, A.; Gordon-Weeks, P.; Hupp, T. DAPK-1 binding to a linear peptide motif in MAP1B stimulates autophagy and membrane blebbing. J. Biol. Chem. 2008, 283, 9999–10014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Maiuri, M.; Galluzi, L.; Morselli, E.; Kepp, O.; Malik, S.; Kroemer, G. Autophagy regulation by p53. Curr. Opin. Cell Biol. 2010, 22, 181185. [Google Scholar] [CrossRef] [PubMed]
  254. Green, D.R.; Kroemer, G. Cytoplasmic functions of the tumour suppressor p53. Nature 2009, 458, 1127–1130. [Google Scholar] [CrossRef] [PubMed]
  255. Comel, A.; Sorrentino, G.; Capaci, V.; Del Sal, G. The cytoplasmic side of p53´s oncosuppressive activities. FEBS Lett. 2014, 588, 2600–2609. [Google Scholar] [CrossRef] [Green Version]
  256. Tasdemir, E.M.; Maiuri, C.; Galluzzi, L.; Vitale, I.; Djavaheri-Mergny, M.; D’Amelio, M.; Criollo, A.; Morselli, E.; Zhu, C.; Harper, F.; et al. Regulation of autophagy by cytoplasmic p53. Nat. Cell Biol. 2008, 10, 676–687. [Google Scholar] [CrossRef] [Green Version]
  257. Tasdemir, E.; Chiara Maiuri, M.; Morselli, E.; Criollo, A.; D’Amelio, M.; Djavaheri-Mergny, M.; Cecconi, F.; Tavernarakis, N.; Kroemer, G. A dual role of p53 in the control of autophagy. Autophagy 2008, 4, 810–814. [Google Scholar] [CrossRef] [Green Version]
  258. Bensaad, K.; Cheung, E.C.; Vousden, K.H. Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO J. 2009, 28, 3015–3026. [Google Scholar] [CrossRef] [Green Version]
  259. Foggetti, G.; Ottaggio, L.; Russo, D.; Mazzitelli, C.; Monti, P.; Degan, P.; Miele, M.; Fronza, G.; Menichini, P. Autophagy induced by SAHA affects mutant P53 degradation and cancer cell survival. Biosci. Rep. 2019, 39, BSR20181345. [Google Scholar] [CrossRef] [Green Version]
  260. Muller, P.A.J.; Vousden, K.H. P53 Mutations in Cancer. Nat. Cell Biol. 2013, 15, 2–8. [Google Scholar] [CrossRef]
  261. Segerba, D.; Bergman, J.; Lambert, J.M.R.; Gorzov, P.; Veprintsev, D.B.; So, M.; Fersht, A.R.; Hainaut, P.; Wiman, K.G.; Bykov, V.J.N. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell 2009, 15, 376–388. [Google Scholar] [CrossRef] [Green Version]
  262. Bykov, V.J.; Zache, N.; Stridh, H.; Westman, J.; Bergman, J.; Selivanova, G.; Wiman, K.G. PRIMA-1(MET) synergizes with cisplatin to induce tumor cell apoptosis. Oncogene 2005, 24, 3484–3491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Bykov, V.J.; Issaeva, N.; Shilov, A.; Hultcrantz, M.; Pugacheva, E.; Chumakov, P.; Bergman, J.; Wiman, K.G.; Selivanova, G. Restoration of the tumor suppressor function to mutant p53 by a low-molecular-weight compound. Nat. Med. 2002, 8, 282–288. [Google Scholar] [CrossRef] [PubMed]
  264. Bykov, V.; Issaeva, N.; Selivanova, G.; Wiman, K.G. Mutant p53-dependent growth suppression distinguishes PRIMA-1 from known anticancer drugs: a statistical analysis of information in the National Cancer Institute database. Carcinogenesis 2002, 23, 2011–2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Foster, B.; Coffey, H.; Morin, M.; Rastinejad, F. Pharmacological rescue of mutant p53 conformation and function. Science 1999, 286, 2507–2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Floquet, C.; Deforges, J.; Rousset, J.-P.; Bidou, L. Rescue of non-sense mutated p53 tumor suppressor gene by aminoglycosides. Nucleic Acids Res. 2011, 39, 3350–3362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Li, D.; Marchenko, N.; Moll, U. SAHA shows preferential cytotoxicity in mutant p53 cancer cells by destabilizing mutant p53 through inhibition of the HDAC6-Hsp90 chaperone axis. Cell Death Diff. 2011, 18, 1904–1913. [Google Scholar] [CrossRef] [Green Version]
  268. Blagosklonny, M.V.; Trostel, S.; Kayastha, G.; Demidenko, Z.N.; Vassilev, L.T.; Romanova, L.Y.; Bates, S.; Fojo, T. Depletion of mutant p53 and cytotoxicity of histone deacetylase inhibitors. Cancer Res. 2005, 65, 7386–7392. [Google Scholar] [CrossRef] [Green Version]
  269. Li, D.; Marchenko, N.D.; Schulz, R. Functional inactivation of endogenous MDM2 and CHIP by HSP90 causes aberrant stabilization of mutant p53 in human cancer cells. Mol. Cancer Res. 2011, 9, 577–588. [Google Scholar] [CrossRef] [Green Version]
  270. Garufi, A.; Pucci, D.; Orazi, V.D.; Cirone, M.; Bossi, G.; Avantaggiati, M.L.; Orazi, G.D. Degradation of mutant p53H175 protein by Zn (II) through autophagy. Cell Death Dis. 2014, 5, e1271. [Google Scholar] [CrossRef]
  271. Yan, W.; Liu, S.; Xu, E.; Zhang, J.; Zhang, Y.; Chen, X.; Chen, X. Histone deacetylase inhibitors suppress mutant p53 transcription via histone deacetylase 8. Oncogene 2013, 32, 599–609. [Google Scholar] [CrossRef] [Green Version]
  272. Hientz, K.; Mohr, A.; Bhakta-guha, D.; Efferth, T. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget 2017, 8, 8921–8946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Alexandrova, E.M.; Yallowitz, A.R.; Li, D.; Xu, S.; Schulz, R.; Proia, D. Improving survival by exploiting tumour dependence on stabilized mutant p53 for treatment. Nature 2015, 523, 352–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Lam, L.; Davis, R.; Pierce, J.; Hepperle, M.; Xu, Y.; Hottelet, M.; Nong, Y.; Wen, D.; Adams, J.; Dang, L.; et al. Small molecule inhibitors of IkB kinase are selectively toxic for subgroups of diffuse large B-cell lymphoma defined by gene expression profiling. Clin. Cancer Res. 2005, 11, 28–40. [Google Scholar] [PubMed]
  275. Nagel, D.; Vincendeau, M.; Eitelhuber, A.C.; Krappmann, D. Mechanisms and consequences of constitutive NF-?B activation in B-cell lymphoid malignancies. Oncogene 2014, 33, 5655–5665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Criollo, A.; Senovilla, L.; Morselli, E.; Vitale, I.; Kepp, O.; Tasdemir, E.; Galluzzi, L.; Shen, S.; Tailler, M.; Delahaye, N.; et al. The IKK complex contributes to the induction of autophagy. EMBO J. 2010, 29, 619–631. [Google Scholar] [CrossRef] [Green Version]
  277. Mortezaee, K.; Najafi, M.; Farhood, B.; Ahmadi, A.; Shabeeb, D.; Musa, A.E. NF-κB targeting for overcoming tumor resistance and normal tissues toxicity. J. Cell. Physiol. 2019, 234, 17187–17204. [Google Scholar] [CrossRef]
  278. Eijkelenboom, A.; Burgering, B. FOXOs: signalling integrators for homeostasis maintenance. Nat. Rev. Mol. Cell Biol. 2013, 14, 83–97. [Google Scholar] [CrossRef]
  279. Kops, G.; Dansen, T.; Polderman, P.; Saarloos, I.; Wirtz, K.; Coffer, P.; Huang, T.; Bos, J.; Medema, R.; Burgering, B. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 2002, 419, 316–321. [Google Scholar] [CrossRef]
  280. Dijkers, P.; Medema, R.; Pals, C.; Banerji, L.; Thomas, N.; Lam, E.; Burgering, B.M.; Raaijmakers, J.A.; Lammers, J.W.; Koenderman, L.; et al. Forkhead transcription factor FKHR-L1 modulates cytokine-dependent transcriptional regulation of p27(KIP1). Mol. Cell. Biol. 2000, 20, 9138–9148. [Google Scholar] [CrossRef] [Green Version]
  281. Dijkers, P.; Medema, R.; Lammers, J.; Koenderman, L.; Coffer, P. Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1. Curr. Biol. 2000, 10, 1201–1204. [Google Scholar] [CrossRef] [Green Version]
  282. Huang, H.; Tindall, D. Dynamic FoxO transcription factors. J. Cell Sci. 2007, 120, 2479–2487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Xiao, D.; Bommareddy, A.; Kim, S.; Sehrawat, A.; Hahm, E.; Singh, S. Benzyl isothiocyanate causes FoxO1-mediated autophagic death in human breast cancer cells. PLoS ONE 2012, 7, e32597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Han, J.; Pan, X.; Xu, Y.; Xiao, Y.; An, Y.; Tie, L.; Pan, Y.; Li, X. Curcumin induces autophagy to protect vascular endothelial cell survival from oxidative stress damage. Autophagy 2012, 8, 812–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Fu, Z.; Tindall, D. FOXOs, cancer and regulation of apoptosis. Oncogene 2008, 27, 2312–2319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Ho, K.; Myatt, S.; Lam, E. Many forks in the path: cycling with FoxO. Oncogene 2008, 27, 2300–2311. [Google Scholar] [CrossRef] [Green Version]
  287. Lin, A.; Piao, H.; Zhuang, L.; Sarbassov, D.; Ma, L.; Gan, B. FoxO transcription factors promote AKT Ser473 phosphorylation and renal tumor growth in response to pharmacological inhibition of the PI3K-AKT pathway. Cancer Res. 2014, 74, 1682–1693. [Google Scholar] [CrossRef] [Green Version]
  288. Bialik, S.; Kimchi, A. The death-associated protein kinases: structure, function and beyond. Annu. Rev. Biochem 2006, 75, 189–210. [Google Scholar] [CrossRef] [Green Version]
  289. Deiss, L.; Feinstein, E.; Berissi, H.; Cohen, O.; Kimchi, A. Identification of a novel serine/threonine kinase and a novel 15-kD protein as potential mediators of the gamma interferon-induced cell death. Genes Dev. 1995, 9, 15–30. [Google Scholar] [CrossRef] [Green Version]
  290. Levin-Salomon, V.; Bialik, S.; Kimchi, A. DAP-kinase and autophagy. Apoptosis 2014, 19, 346–356. [Google Scholar] [CrossRef]
  291. Eisenberg-Lerner, A.; Kimchi, A. DAP-kinase regulates JNK signaling by binding and activating protein kinase D under oxidative stress. Cell Death Differ. 2007, 14, 1908–1915. [Google Scholar] [CrossRef] [Green Version]
  292. Wu, J.; Hu, C.; Gu, Q.; Li, Y.; Song, M. Trichostatin A sensitizes cisplatin-resistant A549 cells to apoptosis by up-regulating death-associated protein kinase. Acta Pharmacol. Sin. 2010, 31, 93–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Komatsu, M.; Kurakawa, H.; Waguri, S.; Taguchi, K.; Kobayashi, A.; Ichimura, Y.; Sou, Y.-S.; Ueno, I.; Sakamoto, A.; Tong, K.I.; et al. The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nat. Cell Biol. 2010, 213–223. [Google Scholar] [CrossRef] [PubMed]
  294. Lau, A.; Wang, X.-J.; Zhao, F.; Villeneuve, N.F.; Wu, T.; Jiang, T.; Sun, Z.; White, E.; Zhang, D.D. A noncanonical mechanism of Nrf2 activation by autophagy deficiency: Direct interaction between Keap1 and p62. Mol. Cell Biol. 2010, 30, 3275–3285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Lahiri, P.; Schmidt, V.; Smole, C.; Kufferath, I.; Denk, H.; Strnad, P.; Rülicke, T.; Fröhlich, L.F.; Zatloukal, K. P62/Sequestosome-1 Is indispensable for maturation and stabilization of Mallory-Denk bodies. PLoS ONE 2016, 11. [Google Scholar] [CrossRef] [PubMed]
  296. Taguchi, K.; Fujikawa, N.; Komatsu, M.; Ishii, T.; Unno, M.; Akaike, T.; Motohashi, H.; Yamamoto, M. Keap1 degradation by autophagy for the maintenance of redox homeostasis. Proc. Natl. Acad. Sci. USA 2012, 109, 13561–13566. [Google Scholar] [CrossRef] [Green Version]
  297. Yang, H.; Ni, H.-M.; Guo, F.; Ding, Y.; Shi, Y.-H.; Lahiri, P.; Fröhlich, L.F.; Rülicke, T.; Smole, C.; Schmidt, V.C.; et al. Sequestosome 1/p62 protein is associated with autophagic removal of excess hepatic endoplasmic reticulum in mice. J. Biol. Chem. 2016, 291, 18663–18674. [Google Scholar] [CrossRef] [Green Version]
  298. Villeneuve, N.F.; Lau, A.; Zhang, D.D. Regulation of the Nrf2-Keap1 antioxidant response by the ubiquitin proteasome system: An insight into cullin-ring ubiquitin ligases. Antioxid. Redox Signal. 2010, 13, 1699–1712. [Google Scholar] [CrossRef] [Green Version]
  299. Bao, L.J.; Jaramillo, M.C.; Zhang, Z.B.; Zheng, Y.X.; Yao, M.; Zhang, D.D.; Yi, X.F. Nrf2 induces cisplatin resistance through activation of autophagy in ovarian carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 1502–1513. [Google Scholar]
  300. Belleza, I.; Giambanco, I.; Minelli, A.; Doanto, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta. Mol. Cell. Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef]
  301. Lu, M.C.; Ji, J.A.; Jiang, Z.Y. The Keap1-Nrf2-ARE pathway as a potential preventive and therapeutic target: An update. Med. Res. Rev. 2016, 36, 924–963. [Google Scholar] [CrossRef]
  302. Wagner, J.M.; Hackanson, B.; Lübbert, M.; Jung, M. Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin. Epigenetics 2010, 1, 117–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Tan, J.; Cang, S.; Ma, Y.; Petrillo, R.L.; Liu, D. Novel histone deacetylase inhibitors in clinical trials as anti-cancer agents. J. Hematol. Oncol. 2010, 3, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. O’Connor, O.; Heaney, M.; Schwartz, L.; Richardson, S.; Willim, R.; MacGregor-Cortelli, B.; Curly, T.; Moskowitz, C.; Portlock, C.; Horwitz, S.; et al. Clinical experience with intravenous and oral formulation of the hovel histone deacetylase inhibitor suberoylanilide hydroxamic acid in patients with advanced hematologic malignancies. J. Clin. Oncol. 2006, 24, 166–172. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview about major effector mechanisms of histone deacetylase (HDAC) inhibitor (HDACi) in cancer cells. Important individually affected molecules (blue font) of the different categories of HDACi- activated anti-tumor pathways (grey boxes) are displayed. HDACi interfere with the deacetylation of histone and non-histone protein deacetylation of many tumor tuppressor genes and oncogenes (among other regulatory proteins) in cancer cells; this posttranslational modification renders them active and thereby induces the different effector mechanisms eventually causing cell death. ↑ upregulation or activation; ↓ downregulation or inhibition; HAT, histone acetyl transferase(s); ROS, rewactive oxygen species; TRX, thioredoxin; TBP2, TRX-binding protein 2; p53, tumor suppressor protein p53; VHL, von Hippel Lindau factor; HIF1α, hypoxia-inducible factor 1α; VEGF, vascular endothelial growth factor; bFGF, basic fibroblast growth factor; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor lappa B; p21, cyclin-dependent kinase inhibitor 1 (p21CIP/WAF1); HDAC6, histon deacetylase 6, c-MYC, cellular homolog of the oncogene (v-myc) of avian myelocytomatosis virus strain 29; c-SRC, cellular SRC kinase oncogene; ER, endoplasmatic reticulum; Ku70, Lupus Ku autoantigen protein p70; Ku 86, Lupus Ku autoantigen protein p86; RAD51 (BRCC5); DNA repair protein RAD51 homolog; BRCA1/2, BRCA1/2, DNA repair associated (prev. breast cancer suceptibility protein ½).
Figure 1. Overview about major effector mechanisms of histone deacetylase (HDAC) inhibitor (HDACi) in cancer cells. Important individually affected molecules (blue font) of the different categories of HDACi- activated anti-tumor pathways (grey boxes) are displayed. HDACi interfere with the deacetylation of histone and non-histone protein deacetylation of many tumor tuppressor genes and oncogenes (among other regulatory proteins) in cancer cells; this posttranslational modification renders them active and thereby induces the different effector mechanisms eventually causing cell death. ↑ upregulation or activation; ↓ downregulation or inhibition; HAT, histone acetyl transferase(s); ROS, rewactive oxygen species; TRX, thioredoxin; TBP2, TRX-binding protein 2; p53, tumor suppressor protein p53; VHL, von Hippel Lindau factor; HIF1α, hypoxia-inducible factor 1α; VEGF, vascular endothelial growth factor; bFGF, basic fibroblast growth factor; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor lappa B; p21, cyclin-dependent kinase inhibitor 1 (p21CIP/WAF1); HDAC6, histon deacetylase 6, c-MYC, cellular homolog of the oncogene (v-myc) of avian myelocytomatosis virus strain 29; c-SRC, cellular SRC kinase oncogene; ER, endoplasmatic reticulum; Ku70, Lupus Ku autoantigen protein p70; Ku 86, Lupus Ku autoantigen protein p86; RAD51 (BRCC5); DNA repair protein RAD51 homolog; BRCA1/2, BRCA1/2, DNA repair associated (prev. breast cancer suceptibility protein ½).
Cancers 12 00109 g001
Figure 2. Different mechanisms of histone deacetylase (HDAC) inhibitor-induced autophagy (grey boxes) and presumed molecular determinants that cause resistance (red font). As many cancer cells already block many pathways leading to apoptosis (black arrow), which cannot be reversed by HDACi treatment, the additional occurrence of resistance to autophagy or the additional use of autophagy inhibitors (red arrow) eventually facilitates cancer cell survival. BECN1, BECLIN-1; Mutp53, mutant p53; MDM2, mouse double minute 2; HSP90, heat shock protein 90; TRX, thioredoxin; MDR1, multiple drug resistance-1.
Figure 2. Different mechanisms of histone deacetylase (HDAC) inhibitor-induced autophagy (grey boxes) and presumed molecular determinants that cause resistance (red font). As many cancer cells already block many pathways leading to apoptosis (black arrow), which cannot be reversed by HDACi treatment, the additional occurrence of resistance to autophagy or the additional use of autophagy inhibitors (red arrow) eventually facilitates cancer cell survival. BECN1, BECLIN-1; Mutp53, mutant p53; MDM2, mouse double minute 2; HSP90, heat shock protein 90; TRX, thioredoxin; MDR1, multiple drug resistance-1.
Cancers 12 00109 g002
Table 1. Mechanisms of HDACi-induced autophagy and potential mechanisms of resistance.
Table 1. Mechanisms of HDACi-induced autophagy and potential mechanisms of resistance.
Autophagic RegulationHDACiCancer Cell TypeRef.Mechanisms of ResistanceRef.
mTOR InhibitionSAHAESS-1[49,153]Constitutive PI3K-AKT signaling
-AKToverexpression
-PI3K mutations
-Loss of PTEN, ARHI, LKB-1
-other upstream factors
Beclin-1 deficiency
Bcl-2 overexpression
SAHAGlioblastoma [136]
Butyrate, SAHAHelaS3 [135,154]
SAHA, OSU-HAD, C42HCC, Hep3B, HepG2 [139][155,156,157,158]
SAHAJurkat T-cells [137]
SAHAGliobastoma SC[138]
ApicidinSalivary MEC[134,159][160,161,162,163]
* MGCD0103Primary CLL [151,152]
* SAHA, TSA, VPA, MS-275, JQ2DS-AMKL cells[64]
ROS AccumulationSAHAJurkat T-cells[137]Increased levels of ROS scavengers/antioxidant enzymes (TRX)[164,165]
SAHACMLL [58]
FK228Gastric carcinoma [165]
M-275HCT116[166]
VPA, SAHAAML (Kasumi-1)[62]
VPA, TSAPaCa44, Panc1[130]
p53 Acetylation & DeficiencySirtinolMCF-7[131]Overexpression/GOF
of mutant p53
Decreased degradation of p53 (MDM2, HSP90)
[167,168,169,170,171]
MHY2256MCF-7 [132]
MHY2256Endometrial [133]
SAHAESS-1[49]
VPA, TSAPaCa44, Panc1[130]
p21 Upregulation SAHA, H40PC-3M, HL-60[142]Deregulated phosphorylation by Akt1
Deregulation by p53
(p53R2; cytoplasmic p21)
[172,173,174,175]
MRJF4PC3[128]
ApicidinSalivary MEC [134,159]
VPA, TSAPaCa44, Panc1[130]
NF-κB Hyper-acetylationSAHA, MS-275PC3[141](Constitutive) NF-κB upregulation[176]
FOXO1 TranscriptionLBH589HepG2, HCT116[145]MDR1 upregulation
Anti-oxidative enzyme upregulation
[177,178,179]
DAPK UpregulationSAHA, TSA, LBH589, JQ2HCT116[147]DAPK hypermethylation & ubiquitination[180,181]
NRF2 upregulationSAHA, TSAHuh-7, MGC-803[146]Aberrant NRF2
expression
[182,183,184]
* Suppression of autophagy due to mTOR activation; HDACi, histone deacetylase inhibitor; SAHA, suberoylanilide hydroxamic acid; VPA, valproic acid; TSA, trichostatin A; MGCD0103, mocetinostat; MS-275, etinostat; FK228, romidepsin; LBH589, panobinostat; SC, stem cells; MEC, Mucoepidermoid carcinoma; CLL, chronic lymphocytic leukemia;; DS-AMKL, down syndrome associated acute myeloid leukemia; AML, acute myeloid leukemia; MDM2, mouse double minute 2; TRX, thioredoxin; HSP90, heat shock protein 90.

Share and Cite

MDPI and ACS Style

Mrakovcic, M.; Fröhlich, L.F. Molecular Determinants of Cancer Therapy Resistance to HDAC Inhibitor-Induced Autophagy. Cancers 2020, 12, 109. https://doi.org/10.3390/cancers12010109

AMA Style

Mrakovcic M, Fröhlich LF. Molecular Determinants of Cancer Therapy Resistance to HDAC Inhibitor-Induced Autophagy. Cancers. 2020; 12(1):109. https://doi.org/10.3390/cancers12010109

Chicago/Turabian Style

Mrakovcic, Maria, and Leopold F. Fröhlich. 2020. "Molecular Determinants of Cancer Therapy Resistance to HDAC Inhibitor-Induced Autophagy" Cancers 12, no. 1: 109. https://doi.org/10.3390/cancers12010109

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

Article Metrics

Back to TopTop