Skip to main content
Biochemistry and Biophysics Reports logoLink to Biochemistry and Biophysics Reports
. 2022 Apr 21;30:101264. doi: 10.1016/j.bbrep.2022.101264

Epigenetic regulation of autophagy in coronavirus disease 2019 (COVID-19)

Hamid Behrouj a, Omid Vakili b, Adel Sadeghdoust c, Neda Aligolighasemabadi d, Parnian Khalili f, Mozhdeh Zamani f, Pooneh Mokarram f,g,
PMCID: PMC9021360  PMID: 35469237

Abstract

The coronavirus disease 2019 (COVID-19) pandemic has become the most serious global public health issue in the past two years, requiring effective therapeutic strategies. This viral infection is a contagious disease caused by new coronaviruses (nCoVs), also called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Autophagy, as a highly conserved catabolic recycling process, plays a significant role in the growth and replication of coronaviruses (CoVs). Therefore, there is great interest in understanding the mechanisms that underlie autophagy modulation. The modulation of autophagy is a very complex and multifactorial process, which includes different epigenetic alterations, such as histone modifications and DNA methylation. These mechanisms are also known to be involved in SARS-CoV-2 replication. Thus, molecular understanding of the epigenetic pathways linked with autophagy and COVID-19, could provide novel therapeutic targets for COVID-19 eradication. In this context, the current review highlights the role of epigenetic regulation of autophagy in controlling COVID-19, focusing on the potential therapeutic implications.

Keywords: COVID-19, Autophagy, Epigenetics, Histone modification, DNA methylation

Graphical abstract

Image 1

Highlights

  • Increasing evidence proposes that autophagy has an essential role in the pathogenicity of SARS-CoV-2.

  • DNA methylation, histone modifications, and non-coding RNAs-dependent regulation are well-known epigenetic mechanisms that regulate cell cycle, cell apoptosis, and the autophagic flux.

  • Deep understanding of the role of epigenetic modifications in the process of autophagy may make us possible to develop potential therapeutic approaches for COVID-19 eradication.

  • The epigenetics-autophagy interplay underlying the COVID-19 is able to improve the understanding of involved mechanisms and identifying novel antiviral therapeutic targets.

Abbreviations:

COVID-19

coronavirus disease 2019

SARS-CoV-2

severe acute respiratory syndrome coronavirus 2

MERS-CoV

Middle East respiratory syndrome coronavirus

WHO

World Health Organization

RBD

receptor-binding domain

ACE2

angiotensin-converting enzyme 2

NRP1

neuropilin-1

TMPRSS2

transmembrane protease serine 2

NSPs

nonstructural proteins

ORFs

open reading frames

RdRp

RNA-dependent RNA polymerase

DMV

double-membrane vesicle

ER

endoplasmic reticulum

ARDS

Acute Respiratory Distress Syndrome

CVD

cardiovascular disease

DM

diabetes mellitus

DIC

disseminated intravascular coagulation

AKI

acute kidney injury

CMA

chaperone-mediated autophagy

Hsc70

heat shock cognate protein 70

LAMP-2AR

lysosomal-associated membrane protein 2A

Atg

autophagy-related genes

ULK1

unc-51-like autophagy activating kinase 1

PAS

phagophore assembly site

PI3K LC3B

phosphoinositide 3-kinase

microtubule-associated protein light chain 3
PE

phosphatidylethanolamine

AMPK

AMP-activated protein kinase

mTORC1

mammalian target of rapamycin complex 1

Akt

protein kinase B

ERK

extracellular signal-regulated kinase

MAPK

mitogen-activated protein kinase

RTCs

replication/transcription complexes

PTMs

post-translational modifications

PKA

protein kinase A

UPR

unfolded protein response

HIF-1α

hypoxia inducible factor 1α

FoxO

forkhead box transcription factor class O

ncRNAs

non-coding RNAs

NOR1

nitro domain-containing protein 1

DAPK

death-associated protein kinase

SOX1

SRY-box transcription factor 1

DNMTs

DNA methyltransferases

ALL

acute lymphatic leukemia

ESCC

esophageal squamous cell carcinoma

H. pylori

Helicobacter pylori

Mtb

Mycobacterium tuberculosis

DRAM1

DNA damage-regulated autophagy modulator protein 1

HATs

histone acetylases

HDACs

histone deacetylases

SIRT

Sirtuin

HIV

human immunodeficiency virus

HMGB1

high-mobility group box 1

miRNAs

MicroRNAs

HCC

hepatocellular carcinoma

α-Syn

alpha-Synuclein

BCG

Bacillus Calmette-Guérin

MEG 3

Maternally expressed gene 3

siRNA

Small interfering RNA

USP22

ubiquitin-specific peptidase 22

1. Introduction

The COVID-19 pandemic in China, and then all over the World, in late December 2019 took everyone by surprise. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) appeared after the Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012 and the severe acute respiratory syndrome coronavirus (SARS-CoV) in 2003 [1]. Unlike the previous two CoV-associated syndromes, the SARS-CoV-2 infection had a higher rate of human-to-human transmission [2]. As a result, the World Health Organization (WHO) finally declared the COVID-19 a global pandemic in March 2020, just a few months after it emerged [3]. SARS-CoV-2, as the causative pathogen for COVID-19, has led to almost 4,010,834 deaths as of July 9, 2021 [4].

SARS-CoV-2 belongs to the family Coronaviridae that are enveloped, positive-sense single-stranded RNA viruses (+ssRNA) [5]. Spike (S) is one of the key structural proteins encoded by almost all coronaviruses, including SARS-CoV-2. S glycoprotein consists of two subunits the N-terminal S1 domain, and the C-terminal S2 domain [6]. The receptor-binding domain (RBD), which is responsible for recognizing the angiotensin-converting enzyme 2 (ACE2) receptors on the target cells, is located on the S1 subunit, while the functional elements required for fusion, such as fusion peptide (FP), two heptad repeats (HR1 and HR2), and the transmembrane [7] domains are located on the S2 subunit [5,6]. The multibasic S1/S2 protease cleavage site is a vital characteristic of SARS-CoV-2 for its viral entry [8]. According to recent evaluations, ACE2, along with neuropilin-1 (NRP1), are the key receptors for the nCoV entry into the host cells [9,10]. After the binding of S proteins to the ACE2 receptors, S proteins need to undergo priming to be further processed and cleaved by the host cell's proteases, such as the human transmembrane protease serine 2 (TMPRSS2), to fuse the viral membrane with the host cell membrane [5]. Following this fusion, SARS-CoV-2 enters the cell by endocytosis. Inside the cytoplasm, the uncoating process results in the release of viral RNA. The genomic material of SARS-CoV-2 acts as a messenger RNA (mRNA), producing structural and nonstructural proteins (NSPs) with the assistance of host ribosomes [11]. The 3′ end of the viral genome encodes four structural proteins, namely spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins. The S protein is responsible for the attachment and fusion of the virus to the host cell receptors. The N protein encapsulates the RNA genome, keeping it stable within the envelope. The 5′ end of the RNA genome consists of two open reading frames (ORFs); ORF1a and ORF1b, which encodes 16 NSPs [5]. NSPs are vital components for viral RNA synthesis. The RNA-dependent RNA polymerase (RdRp) of the virus is the key enzyme of the replication/transcription complex (RTC), formed in a double-membrane vesicle (DMV) by NSPs [12]. For SARS-CoV-2 replication, full-length negative-sense genomic copies are initially created. Next, the aforementioned copies act as templates for the synthesis of the new positive-sense genomic RNA [5]. Finally, after the assembly of newly produced viral RNA and the structural proteins in the endoplasmic reticulum (ER) and Golgi apparatus, new viral particles are released from the infected cells through the exocytosis of secretory vesicles [13].

SARS-CoV-2 transmission is mostly through respiratory aerosols from an infectious person to others. Following the entrance of SARS-CoV-2 into the upper respiratory tract, the virus binds to the ACE-2 receptor, entering the host cell by endocytosis. Within the host cell, the virus goes through replication. Finally, the newly formed viruses are transported via exocytosis into the extracellular space, infecting the other cells [14]. The COVID-19 clinically varies from asymptomatic disease to critical infection. Mild symptoms following the involvement of the upper airways include anosmia, sore throat, dry cough, fever, malaise, and myalgias [15]. In most patients, the virus does not progress beyond the upper respiratory tract because of the sufficient performance of the immune system. However, lower respiratory tract involvement occurs in some individuals, leading to more severe manifestations, including ARDS (defined as Acute Respiratory Distress Syndrome), lymphopenia, and acute cardiac, renal, and hepatic injuries. Furthermore, because of the highly expressed ACE-2 receptors inside the GI tract, especially the small intestine, some patients experience GI symptoms, such as abdominal pain, anorexia, nausea, vomiting, and diarrhea [16]. Complications mainly occurr in patients with severe or critical illness. Age, cardiovascular disease (CVD), chronic lung disease, diabetes mellitus (DM), immunosuppression, and obesity are considered as the major risk factors for COVID-19 complications. In this context, ARDS, acute respiratory failure, sepsis, disseminated intravascular coagulation (DIC), acute liver injury, acute kidney injury (AKI), and pulmonary embolism are some of the commonly seen complications [14].

At present, there is not a definitive cure for SARS-CoV-2 infection. Many clinical trials are trying to identify the most effective drug or combination therapy against the disease. Despite the presence of some suggested treatments to be administered for COVID-19 patients (Table 1), people are still dying from SARS-CoV-2 infection-related complications. Therefore, new therapeutic strategies are urgently needed for controlling the infection to avoid fatal outcomes. For their survival, viruses can manipulate some host cell processes, such as the autophagy machinery [17]. Autophagy is a cellular process that eliminates dysfunctional organelles, misfolded proteins, and intracellular pathogens [18]. Several investigations have attributed a key role in the growth and replication of nCoVs to the autophagic flux [3,19,20]. Additionally, increasing evidence suggests that autophagy has an essential role in the pathogenicity of SARS-CoV-2 [21]. Thus, identifying mechanisms that govern the regulation of autophagy could be therapeutically valuable. Recently, a significant number of studies have reported that epigenetic modifications may regulate the initiation of autophagy [22,23]. DNA methylation, histone modifications, and non-coding RNAs-dependent regulation are well-known epigenetic mechanisms that regulate cell cycle, cell apoptosis, and the autophagic flux [24]. Some epigenetic modifiers also contribute to the regulation of autophagy and potentiate the efficacy of traditional therapeutics [25]. Therefore, gaining an in-depth understanding of the role of epigenetic modifications in the process of autophagy may make it possible to develop potential therapeutic approaches for COVID-19 eradication.

Table 1.

Potential drugs that have been suggested for SARS-CoV-2 infection.

Treatments Mechanisms of Action References
Remdesivir Inhibits viral [26,27]
RNA polymerases
Lopinavir/Ritonavir Inhibit proteases [3]
Favipiravir Inhibits the [28]
RNA-dependent RNA polymerase (RdRp) of RNA viruses
Ivermectin Inhibits the nuclear import of host and viral proteins [29]
Hydroxychloroquine and Chloroquine Prevent endosome trafficking and and viral fusion [30,31]
Arbidol/lopinavir/ritonavir Block the virus entry into the target cells [32,33]
Prezcobix HIV protease inhibitor [34]
Oseltamivir Neuraminidase inhibitor [35]
Tocilizumab Inhibits IL-6 [36,37]
Corticosteroids Decreases inflammation [38]

Considering the lack of knowledge in the field of epigenetic modulation of autophagy during the SARS-CoV-2 infection, the present review will discuss recent findings on the epigenetics-autophagy interplay underlying COVID-19 to improve the understanding of involved mechanisms and identifying novel antiviral therapeutic targets.

2. Interplay between autophagy and COVID-19

2.1. Autophagy: definition and molecular mechanisms

The history of autophagy dates back to early 1960s, when a Belgian scientist, Christian de Duve, introduced the term “autophagy” at the Ciba Foundation Symposium on lysosomes, a discovery that culminated in the 1974 Nobel Prize in Physiology or Medicine [39]. Since then, the physiological and pathophysiological roles of autophagy have been widely investigated. Autophagy (as the programmed cell death type II) is now recognized as a lysosome-dependent controlled degradation process for eliminating unwanted or malfunctioning components [3,40]. In a regulated manner, it enables the breakdown and recycling of cellular components, a key feature of eukaryotic cells, enabling them to renew themselves [41,42]. Although autophagy was previously believed to be a primitive degradation mechanism that is activated to protect the cells from starvation, it has become evident that autophagy can also contribute to the homeostasis of non-starved cells [43]. Additionally, the autophagic flux affects aging, as well as different types of infectious and non-infectious diseases [[44], [45], [46]].

Macroautophagy, microautophagy, chaperone-mediated autophagy (CMA), and crinophagy are the four major subtypes of autophagia that can be differentiated based on the pattern of cytoplasmic components reaching the lysosomal degradative milieu [47]. The term “autophagy” usually refers to macroautophagy except where otherwise stated. Microautophagy sequesters small portions of cytoplasm containing proteins, mainly targeted by heat shock cognate protein 70 (Hsc70), and a small part of the bulk cytoplasm by direct invagination of the lysosomal membrane [48]. In CMA, targeted proteins carrying a specific pentapeptide motif (Lysine-Phenylalanine-Glutamate-Arginine-Glutamine (KFERQ)) are preferentially destroyed by direct transport into the lysosome in a complex with chaperones (e.g. Hsc-70) via the LAMP-2AR (so-called lysosomal-associated membrane protein 2A) [49]. During crinophagy, secretory granules containing excess or obsolete materials, directly combine with late endosomes or lysosomes to eliminate and recycle the leftover secretory material from the cytoplasm [50].

When macroautophagy (hereafter referred to as autophagy) is triggered by intracellular or extracellular stimuli, a double-membrane autophagosome is formed to deliver various materials from the cytoplasm to the lysosome [10]. Autophagosome formation is a complicated process, divided into three consecutive steps, namely initiation, nucleation, and expansion (Fig. 1). This process is monitored by proteins encoded by evolutionarily conserved autophagy-related genes (Atg) [51]. About thirty-six Atg proteins (ATGs) have been identified so far as being critical for the autophagyic flux [52]. During the initiation step of autophagosome formation, the ATG13 binds unc-51-like autophagy activating kinase 1 (ULK1) at the phagophore assembly site (PAS) [53]. Other ATGs are subsequently added to this complex in order to build the ULK1/Atg1 activated unit which consists of ULK1, ATG13, FIP200 (focal adhesion kinase family interacting protein of 200 kD), and ATG101. Then (in nucleation step), ULK1/Atg1 unit activates a class III phosphoinositide 3-kinase (PI3K) complex (Beclin 1, ATG14, VPS15, VPS34) through attaching to the ATG13 [54]. ATG9A, as a transmembrane protein, plays a key role in this step by delivering lipids. ATG9A positive membrane vesicles, correlating with a ATG2-WIPI complex, are shuttled to the site of autophagosome initiation via interacting with the FIP200 [55]. Subsequently, membrane vesicles become fused at the PAS to create a pre-autophagosome structure called the isolation membrane. During the final step (i.e. expansion) of autophagosome formation, the recently created membrane constantly expands to surround and capture cytoplasm and various cellular components (referred to as phagophore) [56]. This step is mediated by two ubiquitin-like systems,: ATG5-ATG12 conjugation step and microtubule-associated protein light chain 3 (LC3B) processing step [57]. Atg4, which is known as a cysteine protease, generates LC3B–I by cleaving LC3B (a cytosolic protein expressed in different cells). LC3B–I is then activated and conjugated with the amino group of the phosphatidylethanolamine (PE) to form LC3B-II [10]. The ATG12–ATG5–ATG16 complex facilitates the recruitment of LC3B-II into both internal and external surfaces of the growing phagophore, where it participates in hemifusion and expansion of membranes, as well as selecting the suitable cargo for elimination [58]. Eventually, a closed bilayer membrane structure, referred to as mature autophagosome, is formed. At this time, the autophagosome will be transferred to the perinuclear region, where the recently formed lysosomes are usually located, to be merged with endosomes or lysosomes to form the autophagolysosome [59].

Fig. 1.

Fig. 1

Schematic description of autophagy (macroautophagy). This process is triggered by several infectious and noninfectious stimuli. Autophagosome formation is divided into three main steps: initiation (ULK1 initiation complex), nucleation (Atg5–Atg12 conjugation, interaction with Atg16L and multi-merization at the phagophore), and expansion (LC3 processing and insertion into the extending phagophore membrane). Then, the autophagosome is fused with the lysosome, and the randomly or selectively captured targets are degraded by lysosomal proteases enzymes.

2.2. The paradoxical role of autophagy in viral infections

Autophagy has been reported to contribute to a vast array of diseases, including bacterial and viral infections, cancers, neurological disorders, and cardiovascular diseases [60,61]. In the last two decades, great efforts have been devoted to studying the complicated interplay between autophagy and CoV infections, and many hypotheses have been proposed to clarify the involved molecular mechanisms. Previous investigations have suggested that autophagy could be involved in direct and indirect antiviral responses such as the clearance of viruses, the presentation of their antigens, the decrease of hyperactive inflammatory responses [41]. In this context, xenophagy, which is also known as virophagy, refers to a kind of selective autophagy, in which specialized autophagy receptors capture the viruses, viral-derived antigens, and neosynthesized viral components [62]. However, viruses can suppress, escape, or utilize the autophagy machinery and ATG proteins of the host cells to replicate or to modify the autophagy to inhibit the cells’ antiviral mechanisms [63,64]. Therefore, the autophagic flux is likely to be a “double-edged sword” in CoV-associated infections, especially the COVID-19. In this regard, the proviral and antiviral roles of autophagy in different viral diseases are summarized in Table 2.

Table 2.

Summary of the proviral/antiviral roles of autophagy in different viral infections.

Viral diseases Proviral/Antiviral role of autophagy References
Classical swine fever (CSF)
Pro-viral: Induction of autophagy to increase viral replication by activation of the PERK and IRE1 pathways
[86]
Crimean-Congo hemorrhagic fever orthonairovirus (CCHFV)
Pro-viral: Virus non-structural protein induces apoptosis by affecting mitochondrial membrane
[87]
Dengue virus (DENV)
Pro-viral: Enhancing autophagy through mTOR signaling molecule
[88]
Human immunodeficiency virus (HIV) Anti-viral: Induction of autophagy by 1α, 25-dihydroxycholecalciferol leads to reduction of HIV replication [89]
Pro-viral: Nef (negative factor) prevents the autophagy process by connecting BECN1 to its inhibitor BCL2
[90]
Hepatitis C virus (HCV) Pro-viral: Decrease in clearance of virus by IFN-α/RBV [91]
Herpes simplex virus type 1(HSV-1)
Anti-viral: MHC I presentation of viral antigen after HSV-1 infection
[92]
Human papillomavirus (HPV)
Pro-viral: Inhibition of autophagy by activation of PI3K/Akt/mTOR signaling lead to virus replication
[93]
Influenza A virus (IAV) Anti-viral: Restriction of IAV infection through inhibiting fusion of virus with endosomes and activation of interferon by ATG16L1 [94]
Pro-viral: IAV induces autophagy which contributes to IAV replication possibly through modulating virus-induced apoptosis
[95]
Vesicular stomatitis virus (VSV) Anti-viral: Representing of viral antigens to Toll-like receptors (TLRs) [96]
Respiratory syncytial virus (RSV)
Pro-viral: Non-structural protein (NS-1) causes viral replication through the mTOR pathway
[97]
Flavivirus
Pro-viral: Replication of virus by NS4A-induced autophagy
[98]
Foot-and-mouth disease virus (FMDV)
Anti-viral: Infection with the foot and mouth disease virus, Atg5-Atg12 boosts the NF–B and IRF3 pathways
[99]
Paramyxovirus
Anti-viral: Inhibiting of virus replication by IFN-induced TDRD7 (Tudor domain containing 7)
[100]
Severe acute respiratory syndrome-coronavirus (SARS-CoV) Pro-viral: Receptor Interacting Protein 3 (Rip3) - mediated oligomerization of SARS causes death cells and promotes SARS CoV infection [101]

2.2.1. Antiviral mechanisms of autophagy in COVID-19

During viral infections, AMP-activated protein kinase (AMPK) suppresses the mammalian target of rapamycin complex 1 (mTORC1) and alternatively triggers PI3K/protein kinase B (Akt1), resulting in autophagy initiation and virion encapsulation. Then, the formation of autolysosomes is activated by these downstream signaling pathways, which are subsequently joined with lysosomes to eliminate its viral contents [65]. However, like most other respiratory CoVs, SARS‐CoV‐2 might have particular mechanisms to escape the autophagy-triggered cellular clearance [66]. Thus, pharmacological targeting of escape mechanisms may mitigate the replication of the virus. Gassen et al. reported in a 2020 preprint that SARS-CoV-2 could inhibit the autophagic flux by interfering with multiple metabolic pathways, including the inhibition of glycolysis by suppressing AMPK and mTORC1 [67], which in turn, pro-autophagic compounds, such as spermidine can block the viral replication. The extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and PI3K/Akt/mTOR, as two signaling responses with inhibitory effects on autophagy, were selectively activated in MERS-CoV-infected hepatocytes [68]. Interestingly, the pharmacological suppression of ERK/MAPK and PI3K/Akt/mTOR pathways led to inhibition of MERS-CoV replication in vitro [68]. Hoffmann et al. (2020) demonstrated that SARS-CoV-2, as well as SARS-CoV-1, used ACE2 as the entry receptor [69,70]. Indeed, ACE2 is a cellular receptor which can suppress cell apoptosis and inhibits autophagy inside the lungs [70]. The HMG-CoA reductase inhibitors, usually known as statins, are a group of drugs commonly used to lower serum cholesterol by reducing its synthesis in the liver [71]. In addition, its well-known lipid-lowering effects, statins have been postulated to possess pleiotropic beneficial actions by regulating numerous biological pathways implicated in antioxidant, anti-inflammatory, or anti-tumor cellular responses [71,72]. Some of the pleiotropic effects attributed to statins may related to their potential role regulating essential proteins involved in autophagy [72,73]. For example, it has been reported that Atorvastatin activated autophagy by upregulating Beclin1 and LC3-II gene and protein expression or via the AMPK/mTOR pathway [74,75]. Accordingly, our recent clinical study revealed that the use of statins in hospitalized COVID-19 patients was linked with a lower risk of death and reduced chance of being subjected to mechanical ventilation [76].

2.2.2. Proviral mechanisms of autophagy in COVID-19

The role of the autophagic flux in viral replication was hypothesized based on the discovery that the induction of double-membrane vesicles (DMVs) formation during the infection process of mouse hepatitis virus (MHV)-infected Vero cells was found to be associated with the LC3 and ATG12 protein, a key maker of autophagosomes formation [77]. Further investigations revealed the co-localization of replicase proteins to cytoplasmic complexes having markers for autophagosome membranes, including LC3, in SARS-CoVs-infected cells [78]. Additionally, it has been found that NSP6, as a key viral replicase protein of Avian CoV (formerly known as infectious bronchitis virus, IBV), is capable of inducing the generation of Atg5 and LC3II-positive autophagosome at the omegasome level [79]. Chen et al. (2014) discovered that the papain-like protease PLP2 (PLP2-TM) of COVs interacts with LC3 and Beclin-1, increasing the autophagosome concentration, preventing autophagosomes from attaching to lysosomes [80]. In another study, it was revealed that NSP6, which has been reported to be expressed in other CoVs, such as SARS-CoV-2, suppresses additional autophagosome expansion and disrupts the virus-to-lysosome transportations [81,82].

Nonetheless, a group of evaluations have challenged the theory of direct involvement of autophagy COVs replication. Consistently, the size of DMVs induced by mammalian CoVs, is reported to be smaller than cellular autophagosomes, and the replication of viruses in MHV-infected cells does not require ATG7 and ATG5, the essential ATGs in the control of autophagosome formation [79,83]. The co-localization of LC3 or green fluorescent protein (GFP)-LC3 with the replication/transcription complexes (RTCs) of SARS-CoV was not observed [84]. This lack of consensus is probably originated from the methodological and/or technical differences of such studies. Another reason for this inconsistency might be that some infectious diseases regulate autophagy in a biphasic, time-dependent manner. For example, Sneha A. Thomas et al. showed that during early infection—and via activation of the Akt pathway—Leishmania actively inhibits the induction of autophagy. However, by 24 h, Leishmania switched from being an inhibitor to an overall inducer of autophagy [85].

3. Epigenetic control of autophagy

Like many other cellular processes, autophagy is regulated by multiple mechanisms. Different signaling pathways, transcriptional processes, post-translational modifications (PTMs), and epigenetic alterations are considered as the most important autophagy regulators [102]. In the case of signaling pathways, the target of rapamycin (TOR) [103] and Ras/cAMP/protein kinase A (PKA) pathways are central to nutrient deprivation-dependent induction of autophagy [104]. It has been demonstrated that TOR down-modulates the autophagic flux, just as the Ras/PKA pathway does [102,105]. Beyond the nutrient signaling, insulin/growth factor pathways [106], AMPK-dependent energy sensing [107], and different types of stress responses such as ER stress [108], oxidative stress [109], and hypoxia [110], which are moderated by unfolded protein response (UPR), antioxidant systems, and hypoxia inducible factor 1α (HIF-1α), respectively, are considered as the other regulating signaling pathways. Yet, Atgs can be also modulated at a transcriptional level, especially when stress occurs. For instance, transcription of the Atg8/LC3, as an autophagosome marker, becomes up-modulated in yeasts and a group of mammalian cells under starvation conditions [102]. Although there is not enough information about the underlying transcriptional factors, forkhead box transcription factor class O (FoxO) is the first transcription factor indicated to be essential for the induction of autophagy according to studies performed on Drosophila larval fat body [111]. Consistent with the aforementioned regulating mechanisms, PTMs critically modulate autophagic flux through modifying a wide variety of ATGs [103,112]. In this context, phosphorylation of autophagy-related complexes, like the Atg1/ULK kinase [113], as well as the acetylation/deacetylation of major ATGs, including ATG5, ATG7, ATG8, and ATG12 [114], are the well-studied PTMs in correlation with autophagy. In spite of the importance of signaling-mediated, transcriptional, and post-transcriptional modulation of autophagy, epigenetic control of this vital process has recently attracted much attention in both physiological and pathological/infectious conditions [115]. DNA methylation/demethylation, histone modifications, and non-coding RNAs (ncRNAs)-mediated regulation are categorized as the most prominent epigenetic alterations, which are highlighted below.

3.1. DNA methylation/demethylation

DNA methylation is a dynamic process, in which methylated targets can be subsequently demethylated [116]. Several Atgs have been reported to be methylated and suppressed to block autophagy in particular pathological conditions. Atg1/ULK kinase, Beclin1/Atg6, LC3/Atg8, and lysosomal-associated membrane protein 2 (LAMP2) gene are some examples in this field [[116], [117], [118]]. Moreover, genes encoding autophagy regulatory molecules, such as nitro domain-containing protein 1 (NOR1), death-associated protein kinase (DAPK), and SRY-box transcription factor 1 (SOX1) can also be modified by DNA methylation [116].

DNA methylation generally takes place at three major levels: (i) loss of imprinting (LOI), (ii) hypomethylation, and (iii) hypermethylation, in which DNA hypermethylation has a more significant role in the regulation of autophagy than the two others [119,120]. In this context, DNA methyltransferases (DNMTs) have been reported to have vital roles in epigenetic control of autophagyic flux, in part because the improper methylation of Atgs, which results in changes in ATG expression levels, was found to be correlated with a variety of human diseases [121]. For example, children with acute lymphoblastic leukemia (ALL) were found to overexpress DNMT1 compared to healthy individuals, which is the main cause of hypermethylation of the ATG5 and LC3B gene promoters [122]. It was also revealed that improper methylation of LC3Av1, as a member of the LC3 gene family, could be in close association with suppressed autophagy in esophageal squamous cell carcinoma (ESCC) cells, as well as tumorigenesis [123]. Klotho (KL) and protocadherin 17 (PCDH17) tumor suppressor genes, expressed in colorectal and gastric cancer cells, respectively, are other genes that play a role in inducing autophagy. Interestingly, these genes can go under promoter methylation, and thus be employed as epigenetic diagnostic markers for the aforementioned tumors [124,125]. The overexpressed PCDH17 can stimulate autophagy, accompanied by the elevation of autophagy vacuoles and up-modulation of particular ATGs, such as ATG12 and ATG5 [124].

Beyond the cancers, DNA methylation can also orchestrate infectious circumstances. In this context, Helicobacter pylori (H. pylori)-induced DNA methylation has been reported to modulate several Atgs/ATGs in infected gastric mucosae [126,127]. MAP1LC3Av1 is one of these genes that become down-modulated following the hypermethylation in H. pylori-infected cancerous tissues, while it is not expressed in H. pylori negative tissues. Muhammad et al. interestingly showed that MAP1LC3Av1 suppression could disrupt the autophagic flux, resulting in an elevation in tumorigenicity of gastric epithelial cells [128]. Mycobacterium tuberculosis (Mtb) is another pathogen, which provokes its survival in host cells trough inhibiting the autophagy. Indeed, Mtb blocks autophagy in an mTOR-independent manner by triggering the hypermethylation of histones at the Atg5 and Atg7 promoters trough stimulation of p38-MAPK- and EHMT2 methyltransferase-dependent signaling cascades [129].

It should be noted that DNA demethylation can also contribute to the epigenetic control of autophagic flux. TET enzymes (identified as ten-eleven translocation family) are the key contributors to DNA demethylation [130]. Among all TETs, TET1 has been reported to be more often associated with autophagy, as it can exert its tumor suppressive effects. Further, CpG demethylase activities through operation of autophagy, especially at early stages [131]. TET1 can also modulate some Atgs, namely Atg13 and DRAM1 (defined as DNA damage-regulated autophagy modulator protein 1) in cancerous cells [132]. Nonetheless, other TETs may regulate autophagy as well. TET2 is a good example in this field, which can modulate autophagy by affecting the methylation of BECN1 promoter in atherosclerotic conditions [133]. In sum, more investigations should be performed to determine the exact interplay between TET enzymes and autophagic flux.

3.2. Histone acetylation/deacetylation

A group of evaluations have been recently focused on the role of histone acetylases (HATs), as well as histone deacetylases (HDACs) in the epigenetic modulation of autophagy [120,134]. Lysine acetyltransferases (KATs), such as KAT3A, KAT3B, and KAT5, are a major group of HATs, established in the control of autophagy. In addition, Sirtuin 1-3 (SIRT1-3), which are classified as key HDAC family members, are also involved in the modulation of autophagic flux [134]. The H4K16ac, a well-known acetylated histone, has a special effect on transcriptional modulation of Atgs and also stimulates a feedback route that regulates the induction of autophagy. Füllgrabe et al. demonstrated that SIRT1 and KAT8/human males, absent in the first (hMOF)/MYST1, affected the acetylation of H4K16, resulting in a histone shift and subsequent modulation of autophagy [135,136]. Down-modulation of Drosophila MOF and H4K16ac is also correlated with rapamycin/starvation-induced autophagy. More interestingly, SIRT1 can be considered as a limiting factor for autophagy inside the nucleus, where H4K16 deacetylation could repress different ATGs [22]. This fact is analogous to stimulating role of SIRT1 inside the cytoplasm, which deacetylates key ATGs (e.g. ATG5, ATG7, etc.) [22]. Accordingly, if H4K16ac deacetylation is suppressed as a result of SIRT1 deactivation or hMOF up-regulation, the autophagy might be triggered, and cell death would occur. Indeed, it is the H4K16 acetylation status that determines the autophagy-dependent cell death or survival [135,137].

In the case of infectious diseases, there is a well-studied crosstalk between HDAC-mediated regulation of autophagy and human immunodeficiency virus (HIV) infection [138]. Demonté et al. showed that HDAC inhibitors can trigger autophagy-mediated degradation of HIV particles; a valuable finding that reveals the therapeutic capacity of these inhibitors to ameliorate the HIV infection [139]. HDAC inhibition can also be involved in amplification of antimicrobial responses against Mtb by regulating the autophagic flux in a manner that is not fully understood [140,141]. Hepatitis B is another example in this field. HBx, a HBV X protein, is a key modulator of HBV-induced autophagy. Nonethelesss, the cytoplasmic high-mobility group box 1 (HMGB1) is an autophagy inducer, whose cytoplasmic translocation is strongly associated with its acetylation status [142]. Fu et al. demonstrated that HBx up-regulated the HMGB1 and stimulated its cytoplasmic translocation trough acetylation in order to accelerate autophagy. They also identified that nuclear HDAC could trigger HBx-induced hyperacetylation, leading to HMGB1 translocation, mediated by HDAC1 isoform [142]. Moreover, cytoplasmic HBx-to-HMGB1 attachment can initiate autophagy inside the hepatocytes [142]. However, further evaluations are definitely needed to reveal the exact interaction between histone acetylation/deacetylation and the process of autophagy in various infections.

3.3. Non-coding RNA-mediated modifications

In mammals, just like the other eukaryotes, the transcription process mostly results in the production of ncRNAs [120,143]. NcRNAs are believed to be responsible for a wide variety of biological functions. For instance, they can protect genomes during genome rearrangement or the process of DNA synthesis [144,145]. Recent evaluations have indicated that ncRNAs can also be involved in the modulation of autophagy, in which they contribute to the pathogenesis of multiple diseases [146].

MicroRNAs (miRNAs) are a major subgroup of ncRNAs, participating in the control of autophagy [120]. Indeed, miRNAs have been reported to be highly conserved molecules, which can epigenetically modulate the autophagic flux in multiple stages [147]. In this regard, miR-7, miR-101, miR-20a, miR-25,miR-106b, miR-199a, miR-17−5p, miR-595, miR-4487, and miR-409−3p are some examples of autophagy modulators [148]. The function of these miRNAs on autophagy is summarized in Table 3. MiR-7, as a tumor suppressor miRNA, has the ability to increase autophagy in hepatocellular carcinoma (HCC) cells; Wang et al. declared that miR-7-mediated induction of autophagy was a consequence of mTOR inhibition [149]. Furthermore, miR-101 has been demonstrated to be a potential autophagy inhibitor in HCC and breast cancer cells through targeting ATG4D, RAB5A, and stathmin1 [150]. In multiple system atrophy (MSA), miR-101 down-modulation can trigger the autophagic flux to mitigate the accumulation of oligodendroglial alpha-Synuclein (α-Syn) [151]. Further analyses on breast cancer cells revealed another miRNA as an autophagy regulator, miR-25, which modulates autophagy in correlation with cancer drug resistance. It has been confirmed that miR-25 silencing can stimulate autophagic cell death through increasing the expression of ULK1 [152]. Some of the aforementioned miRNAs can also regulate autophagy in infectious conditions [153]. For instance, miR-17-5p has been shown to be a positive modulator of autophagy in Mtb infection through interacting with Beclin-1 and Mcl-1 [154]. On the contrary, Bacillus Calmette-Guérin (BCG) infection-induced overexpression of miR-17-5p blocks the initiation of autophagy and increases the growth of Mycobacteria inside the cells [155]. MiR-106b is another miRNA in this field, which is reported to play a role in the operation of cathepsin S activity, as a crucial step in phagosomal acidification and the subsequent removal of bacteria [156]. However, miR-106b can also affect other autophagic markers such as ATG16L1 in conditions other than microbial infections [156,157].

Table 3.

Autophagy-related microRNAs (miRNAs).

miRNAs Effect on autophagy References
miR-7
Activation: via mTOR inhibition
[173]
miR-101
Inhibition: through targeting ATG4D, RAB5A, and stathmin1
[150,174]
miR-20a Inhibition: via targeting FIP200 [175]
miR-25 Inhibition: through targeting ULK1 [176]
miR-106b
Inhibition: through targeting ATG16L1
[177]
miR-199a Inhibition: via targeting ATG7 [178]
miR-17−5p Inhibition in cancer cells: via targeting BECN1 [179]
Activation in Mycobacterium tuberculosis condition: through targeting Mcl-1 and STAT3 [154]
Inhibition in Bacillus Calmette-Guérin (BCG) condition: through targeting ULK1
[155]
miR-595
Inhibition: through targeting ULK1
[180]
miR-4487
Inhibition: through targeting ULK1
[180]
miR-409−3p Inhibition: through targeting BECN1 [181]

In addition to miRNAs, long non-coding RNAs (lncRNAs) have also been reported to interact with autophagy as unusual modulators [120]. LncRNAs affect the autophagy process through mediation of Atg expression levels, as well as interfering with DNA, RNA, and/or protein molecules [145,158,159]. Maternally expressed gene 3 (MEG 3), which is a tumor suppressor lncRNA, is inversely correlated with the autophagosome marker MAP1A/MAP1B-LC3/ATG8. Small interfering RNA (siRNA)-mediated inhibition of MEG3 was indicated to contribute to autophagy initiation, cell proliferation, and repression of apoptotic flux in bladder cancer cells [160]. Furthermore, the suppressed MEG3 in BCG-infected macrophages, accelerates the clearance of Mycobacterium bovis BCG through stimulation of autophagy [161]. HULC, as another lncRNA overexpressed in HCC, can indirectly induce the process of autophagy through up-regulating the ubiquitin-specific peptidase 22 (USP22) following the suppression of miR-6886−3p, miR-6825−5p, and miR-6845−5p. Xiong et al. declared that USP22 actually restored SIRT1, and inhibited the ATG5/ATG7 acetylation to trigger the autophagic flux [162].

Circular RNAs (circRNAs) are another form of noncoding RNAs that are usually expressed in tissues and body fluids [163]. The circRNAs are reported to have diverse functions in mammals and plants. They can act as microRNA (miRNA) sponges to regulate the function of mRNA [164]. CircRNAs can also play a role in RNA-binding protein (RBP) assembly or allosteric regulation by RBPs to affect biological functions [164]. The circRNAs play essential roles in human diseases and are considered to be ideal biomarkers of these diseases due to their high stability [165]. Evidence has shown that circRNAs influence the course of a disease by regulating autophagy [166]. For example, following binding with MIR139-5p, circEIF3K inhibits the functions of MIR139-5p, the phosphorylation of MAPK/Erk, and the expression of BCL2, which promotes the autophagy in somatic inflammatory cells of patients with tubal inflammation, thereby inhibiting cell vitality and promoting apoptosis [167]. circCDK8 enhances autophagy in periodontal ligament stem cells during hypoxia via upregulation of ATG5 [168]. mmu_circ_0000250 promotes autophagy in endothelial progenitor cells through upregulation of SIRT1 expression [3]. Furthermore, mmu_circ_0000623 inhibits liver fibrosis via activation of cell autophagy [169]. In lung fibroblasts, circ012091 inhibits the expression of the autophagy-associated protein LC3-II by inhibiting the expression of PPP1R13B, a major pro-apoptotic protein of the TP53 family, thus inhibiting autophagy in the cells [170]. During inflammatory bowel disease, circPabpn1 attenuates autophagy in intestinal epithelial cells via downregulation of Atg16l1 gene [171]. Additionally, Virus-derived circRNAs and/or differentially-expressed host circular RNAs have been observed following various virus infections. For instance, following infection with influenza Virus, circGATAD2A increases viral replication by inhibiting autophagy [172]. Although, the role of circRNAs in autophagy regulation has been investigated in various diseases, currently, there is no study about the modulation of autophagy by circRNAs in COVID-19 cases. Therefore, future studies should focus on identifying circRNAs that regulate autophagy in COVID-19 patients because they could be useful in understanding, detecting, and treating COVID-19.

In sum, these findings show that epigenetic modifications play an important role in the control of autophagy during various diseases.

4. Targeting autophagy through epigenetic modulations as a novel therapeutic strategy against COVID-19

Given the important role of autophagy in COVID-19 exacerbations and also regarding the essential role of epigenetic mechanisms in autophagy regulation, epigenetic therapeutics are promising strategies to modify autophagy during the COVID-19. As mentioned above, the histone modification and DNA methylation are commonly evaluated and broadly occurring molecular mechanisms that control the autophagic flux. Relevant analyses in this regard have focused on the significance of inhibitors of these epigenetic alterations to modulate the autophagy. To date, FDA has approved multiple DNMT and HDAC inhibitors for therapeutic purposes. 5-Azacytidine and 5-aza-2′-deoxycytidine (decitabine) are frequently used medications to stimulate DNA de-methylation by incorporation into DNA. Vorinostat (SAHA), Belinostat, romidepsin, trichostatin A (TSA), and Panobinostat are commonly used drugs for HDAC inhibition. Some of these drugs are already used for the treatment of several cancers through autophagy modulation (Table 4). For instance, 5-Azacytidine and decitabine activate autophagy in leukemic T cells through enhancement of the ROS generation, as well as the up-regulation of ATGs [182,183]. Similarly, SAHA induces autophagy in MDA-MB-231, DLD1, T cell lymphoma HUT78, and colorectal cancer cells via p53 degradation and up-regulation of LC3-II and ATG5 [[184], [185], [186]]. Nevertheless, it was reported that SAHA suppresses autophagy in MCF7 breast cancer cells through down-regulation of p62 and overexpression of LC3-II and Beclin-1 [187]. Romidepsin has been shown to be capable of inducing the autophagy in gastric tumor cells through the activation of MAPK pathway and increasing the ROS levels [188,189]. Furthermore, TSA activates the autophagic flux in HCT116 and HepG2 cancer cells by inhibiting mTOR and inducing FOXO1 transcription factor [155]. Panobinostat also up-regulates autophagy in Human pre-B ALL Nalm-6 cells via the activation of FOXO3 and FOXO4 transcription factors [190]. In addition to anti-cancer effects, several studies reported that epigenetic medications, such as DNMT1 and HDAC inhibitors, potentially inhibit SARS-CoV-2 activities [191]. For example, SAHA was reported to be responsible for decreasing the replication of initiating genes, as well as the stimulation of cellular proteins in charge of viral inhibition [192]. VPA was demonstrated to be involved in minimal modulation of SARS-CoV-2 growth [193]. Therefore, common antiviral drugs, e.g., remdesivir, ribavirin, favipiravir, and galidesivir, could be preferentially used in combination with DNMT inhibitors, such as decitabine, and azacitidine, or HDAC inhibitors like vorinostat, belinostat, panobinostat, and TSA. Nonetheless, more preclinical analyses, as well as clinical trials are needed to confirm the clinical benefits of the corresponding combined candidates.

Table 4.

Epigenetic regulators associated with autophagy.

Inhibitors Effect on autophagy References
5-Azacytidine
Up-regulation
[182]
decitabine
Up-regulation
[183]
Zebularine
Up-regulation
[194]
Vorinostat (SAHA)
Up-regulation
[[184], [185], [186]]
Romidepsin
Up-regulation
[188,189]
Trichostatin A (TSA)
Up-regulation
[155]
Tucidinostat Down-regulation [195]
Panobinostat
Up-regulation
[190]
Givinostat
Down-regulation
[196]
Mocetinostat Down-regulation [197]

5. Conclusion and future perspective

As it is highlighted in the current review, autophagy has been markedly implicated in SARS-CoV-2 infection. Today, we know that epigenetic control of autophagy is an important regulatory step in various physiological and pathological conditions, as epigenetic dysregulation of Atg/ATGs is significantly involved in the etiology of different infectious and non-infectious diseases (e.g., cancers, metabolic disorders, and neurodegenerative diseases). However, epigenetic regulation of autophagy in COVID-19 has not significantly been reported. It is therefore imperative to understand the role of epigenetic mechanisms in the regulation of autophagy in COVID-19 because the application of epigenetic modulators, such as demethylating agents or HDAC inhibitors, not only aims to normalize atypical epigenetic patterns on DNA sequences or histones but also provides a newer therapeutic opportunity to regulate autophagy in COVID-19 patients.

Funding sources

This project was supported by the National Institute for Medical Research Development (NIMAD) Grant No: 943267.

Declaration of competing interest

All authors have no conflicts of interest to declare.

Acknowledgments

This project was supported by the National Institute for Medical Research Development (NIMAD) grant No: 943267. All authors contributed substantially to the design and conception of the study, writing the manuscript.

Data availability

Data will be made available on request.

References

  • 1.Ramírez Hernández E., et al. The role of the SARS-CoV-2 S-protein glycosylation in the Interaction of SARS-CoV-2/ACE2 and immunological responses. Viral Immunol. 2021;34(3):165–173. doi: 10.1089/vim.2020.0174. [DOI] [PubMed] [Google Scholar]
  • 2.Majumder J., Minko T. Recent developments on therapeutic and diagnostic approaches for COVID-19. AAPS J. 2021;23(1):1–22. doi: 10.1208/s12248-020-00532-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Shi R., et al. Exosomes derived from mmu_circ_0000250-modified adipose-derived mesenchymal stem cells promote wound healing in diabetic mice by inducing miR-128-3p/SIRT1-mediated autophagy. Am. J. Physiol. Cell Physiol. 2020;1;318(5):C848–C856. doi: 10.1152/ajpcell.00041.2020. [DOI] [PubMed] [Google Scholar]
  • 4.Megha K., Nayar S.A., Mohanan P. Vaccine and vaccination as a part of human life: in view of Covid‐19. Biotechnol. J. 2021 doi: 10.1002/biot.202100188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.V’kovski P., et al. Coronavirus biology and replication: implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021;19(3):155–170. doi: 10.1038/s41579-020-00468-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mahmoud I.S., et al. SARS-CoV-2 entry in host cells-multiple targets for treatment and prevention. Biochimie. 2020;175:93–98. doi: 10.1016/j.biochi.2020.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rybak-Wolf A., et al. Circular RNAs in the mammalian brain are highly abundant, conserved, and dynamically expressed. Mol. Cell. 2015;58(5):870–885. doi: 10.1016/j.molcel.2015.03.027. [DOI] [PubMed] [Google Scholar]
  • 8.Lemmin T., et al. Structures and dynamics of the novel S1/S2 protease cleavage site loop of the SARS-CoV-2 spike glycoprotein. J. Struct. Biol. X. 2020;4 doi: 10.1016/j.yjsbx.2020.100038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cantuti-Castelvetri L., et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 2020;370(6518):856–860. doi: 10.1126/science.abd2985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Siri M., et al. Autophagy, unfolded protein response, and neuropilin-1 cross-talk in SARS-CoV-2 infection: what can Be learned from other coronaviruses. Int. J. Mol. Sci. 2021;22(11):5992. doi: 10.3390/ijms22115992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Romano M., et al. A structural view of SARS-CoV-2 RNA replication machinery: RNA synthesis, proofreading and final capping. Cells. 2020;9(5):1267. doi: 10.3390/cells9051267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Van Hemert M.J., et al. SARS-coronavirus replication/transcription complexes are membrane-protected and need a host factor for activity in vitro. PLoS Pathog. 2008;4(5) doi: 10.1371/journal.ppat.1000054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Marian A.J. Current state of vaccine development and targeted therapies for COVID-19: impact of basic science discoveries. Cardiovasc. Pathol. 2021;50 doi: 10.1016/j.carpath.2020.107278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Parasher A. COVID-19: current understanding of its pathophysiology, clinical presentation and treatment. Postgrad. Med. 2021;97(1147):312–320. doi: 10.1136/postgradmedj-2020-138577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gandhi R.T., Lynch J.B., Del Rio C. Mild or moderate covid-19. N. Engl. J. Med. 2020;383(18):1757–1766. doi: 10.1056/NEJMcp2009249. [DOI] [PubMed] [Google Scholar]
  • 16.Berlin D.A., Gulick R.M., Martinez F.J. Severe covid-19. N. Engl. J. Med. 2020;383(25):2451–2460. doi: 10.1056/NEJMcp2009575. [DOI] [PubMed] [Google Scholar]
  • 17.Dreux M., Chisari F.V. Viruses and the autophagy machinery. Cell Cycle. 2010;9(7):1295–1307. doi: 10.4161/cc.9.7.11109. [DOI] [PubMed] [Google Scholar]
  • 18.Mizushima N. Autophagy: process and function. Genes Dev. 2007;21(22):2861–2873. doi: 10.1101/gad.1599207. [DOI] [PubMed] [Google Scholar]
  • 19.Bonam S.R., et al. Autophagy as an emerging target for COVID-19: lessons from an old friend, chloroquine. Autophagy. 2020;16(12):2260–2266. doi: 10.1080/15548627.2020.1779467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Miller K., et al. Coronavirus interactions with the cellular autophagy machinery. Autophagy. 2020;16(12):2131–2139. doi: 10.1080/15548627.2020.1817280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.García-Pérez B.E., et al. Taming the autophagy as a strategy for treating COVID-19. Cells. 2020;9(12):2679. doi: 10.3390/cells9122679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Füllgrabe J., Klionsky D.J., Joseph B. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat. Rev. Mol. Cell Biol. 2014;15(1):65–74. doi: 10.1038/nrm3716. [DOI] [PubMed] [Google Scholar]
  • 23.Shin H.-J.R., et al. Epigenetic and transcriptional regulation of autophagy. Autophagy. 2016;12(11):2248–2249. doi: 10.1080/15548627.2016.1214780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kim J., Samaranayake M., Pradhan S. Epigenetic mechanisms in mammals. Cell. Mol. Life Sci. 2009;66(4):596–612. doi: 10.1007/s00018-008-8432-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sui X., et al. Epigenetic modifications as regulatory elements of autophagy in cancer. Cancer Lett. 2015;360(2):106–113. doi: 10.1016/j.canlet.2015.02.009. [DOI] [PubMed] [Google Scholar]
  • 26.Wang Y., et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395(10236):1569–1578. doi: 10.1016/S0140-6736(20)31022-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Grein J., et al. Compassionate use of remdesivir for patients with severe Covid-19. N. Engl. J. Med. 2020;382(24):2327–2336. doi: 10.1056/NEJMoa2007016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cai Q., et al. Experimental treatment with favipiravir for COVID-19: an open-label control study. Engineering. 2020;6(10):1192–1198. doi: 10.1016/j.eng.2020.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Caly L., et al. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antivir. Res. 2020;178 doi: 10.1016/j.antiviral.2020.104787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chen J., et al. A pilot study of hydroxychloroquine in treatment of patients with moderate COVID-19. J. Zhejiang Univ. (Med. Sci.) 2020;49(2):215–219. doi: 10.3785/j.issn.1008-9292.2020.03.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gautret P., et al. Clinical and microbiological effect of a combination of hydroxychloroquine and azithromycin in 80 COVID-19 patients with at least a six-day follow up: a pilot observational study. Trav. Med. Infect. Dis. 2020;34 doi: 10.1016/j.tmaid.2020.101663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Huang J.-L., Su M., Wu D.-P. Functional roles of circular RNAs in Alzheimer's disease. Ageing Res. Rev. 2020;60 doi: 10.1016/j.arr.2020.101058. [DOI] [PubMed] [Google Scholar]
  • 33.Zhu Z., et al. Arbidol monotherapy is superior to lopinavir/ritonavir in treating COVID-19. J. Infect. 2020;81(1):e21–e23. doi: 10.1016/j.jinf.2020.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhang Q., et al. Clinical trial analysis of 2019‐nCoV therapy registered in China. J. Med. Virol. 2020;92(6):540–545. doi: 10.1002/jmv.25733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chen N., et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507–513. doi: 10.1016/S0140-6736(20)30211-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Luo P., et al. Tocilizumab treatment in COVID‐19: a single center experience. J. Med. Virol. 2020;92(7):814–818. doi: 10.1002/jmv.25801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Guo T., et al. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol. Neurodegener. 2020;15(1):1–37. doi: 10.1186/s13024-020-00391-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Huang C., et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.De Duve C., Wattiaux R. Functions of lysosomes. Annu. Rev. Physiol. 1966;28(1):435–492. doi: 10.1146/annurev.ph.28.030166.002251. [DOI] [PubMed] [Google Scholar]
  • 40.Behrouj H., et al. Autophagy flux correlates with upregulation of AKT-1 in RAS mutated colon cancer cells. Middle East J. Cancer. 2021;12(4):457–465. [Google Scholar]
  • 41.Carmona-Gutierrez D., et al. Digesting the crisis: autophagy and coronaviruses. Microb. Cell. 2020;7(5):119. doi: 10.15698/mic2020.05.715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Behrouj H., et al. Effect of casein kinase 1α inhibition on autophagy flux and the AKT/phospho-β-catenin (S552) axis in HCT116, a RAS-mutated colorectal cancer cell line. Can. J. Physiol. Pharmacol. 2021;99(3):284–293. doi: 10.1139/cjpp-2020-0449. [DOI] [PubMed] [Google Scholar]
  • 43.Kobayashi S. Choose delicately and reuse adequately: the newly revealed process of autophagy. Biol. Pharm. Bull. 2015;38(8):1098–1103. doi: 10.1248/bpb.b15-00096. [DOI] [PubMed] [Google Scholar]
  • 44.Mokarram P., et al. Autophagy and cancer research in Iran. Autophagy. 2019;15(11):2039–2042. doi: 10.1080/15548627.2019.1606638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Khandia R., et al. A comprehensive review of autophagy and its various roles in infectious, non-infectious, and lifestyle diseases: current knowledge and prospects for disease prevention, novel drug design, and therapy. Cells. 2019;8(7):674. doi: 10.3390/cells8070674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ren J., Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol. Sci. 2018;39(12):1064–1076. doi: 10.1016/j.tips.2018.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rajak S., et al. MTORC1 inhibition drives crinophagic degradation of glucagon. Mol. Metabol. 2021;53 doi: 10.1016/j.molmet.2021.101286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mannack L.V., Lane J.D. The autophagosome: current understanding of formation and maturation. Res. Rep. Biochem. 2015;5:39–58. [Google Scholar]
  • 49.Dong S., et al. Chaperone-mediated autophagy sustains haematopoietic stem-cell function. Nature. 2021;591(7848):117–123. doi: 10.1038/s41586-020-03129-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Csizmadia T., Juhász G. Crinophagy mechanisms and its potential role in human health and disease. Prog. Mol. Biol. Transl. Sci. 2020;172:239–255. doi: 10.1016/bs.pmbts.2020.02.002. [DOI] [PubMed] [Google Scholar]
  • 51.Klionsky D.J. Taylor & Francis; 2012. Look People,“Atg” Is an Abbreviation for “autophagy-related.” That's it. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Subramani S., Malhotra V. Non‐autophagic roles of autophagy‐related proteins. EMBO Rep. 2013;14(2):143–151. doi: 10.1038/embor.2012.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Li Z., et al. ULK1-ATG13 and their mitotic phospho-regulation by CDK1 connect autophagy to cell cycle. PLoS Biol. 2020;18(6) doi: 10.1371/journal.pbio.3000288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Russell R.C., et al. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat. Cell Biol. 2013;15(7):741–750. doi: 10.1038/ncb2757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Maeda S., et al. Structure, lipid scrambling activity and role in autophagosome formation of ATG9A. Nat. Struct. Mol. Biol. 2020;27(12):1194–1201. doi: 10.1038/s41594-020-00520-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mehrbod P., et al. The roles of apoptosis, autophagy and unfolded protein response in arbovirus, influenza virus, and HIV infections. Virulence. 2019;10(1):376–413. doi: 10.1080/21505594.2019.1605803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yu L., Chen Y., Tooze S.A. Autophagy pathway: cellular and molecular mechanisms. Autophagy. 2018;14(2):207–215. doi: 10.1080/15548627.2017.1378838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Li X., He S., Ma B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer. 2020;19(1):1–16. doi: 10.1186/s12943-020-1138-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chew L.H., et al. Structural characterization of the Saccharomyces cerevisiae autophagy regulatory complex Atg17-Atg31-Atg29. Autophagy. 2013;9(10):1467–1474. doi: 10.4161/auto.25687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jiang P., Mizushima N. Autophagy and human diseases. Cell Res. 2014;24(1):69–79. doi: 10.1038/cr.2013.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Levine B., Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42. doi: 10.1016/j.cell.2007.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Orvedahl A., et al. Image-based genome-wide siRNA screen identifies selective autophagy factors. Nature. 2011;480(7375):113–117. doi: 10.1038/nature10546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mao J., et al. Autophagy and viral infection. Autophagy Regul. Innate Immun. 2019:55–78. doi: 10.1007/978-981-15-0606-2_5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gorshkov K., et al. The SARS-CoV-2 Cytopathic Effect Is Blocked by Lysosome Alkalizing Small Molecules. ACS Infectious Diseases. 2021;7(6):1389–1408. doi: 10.1021/acsinfecdis.0c00349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tang S.-W., et al. Impact of cellular autophagy on viruses: insights from hepatitis B virus and human retroviruses. J. Biomed. Sci. 2012;19(1):1–11. doi: 10.1186/1423-0127-19-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Brest P., et al. Open questions for harnessing autophagy-modulating drugs in the SARS-CoV-2 war: hope or hype? Autophagy. 2020;16(12):2267–2270. doi: 10.1080/15548627.2020.1779531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gassen N.C., et al. SARS-CoV-2-mediated dysregulation of metabolism and autophagy uncovers host-targeting antivirals. Nature Communications. 2021;12:1–15. doi: 10.1038/s41467-021-24007-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kindrachuk J., et al. Antiviral potential of ERK/MAPK and PI3K/AKT/mTOR signaling modulation for Middle East respiratory syndrome coronavirus infection as identified by temporal kinome analysis. Antimicrob. Agents Chemother. 2015;59(2):1088–1099. doi: 10.1128/AAC.03659-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hoffmann M., et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271–280. doi: 10.1016/j.cell.2020.02.052. e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li Y., et al. Angiotensin-converting enzyme 2/angiotensin-(1–7)/Mas axis prevents lipopolysaccharide–induced apoptosis of pulmonary microvascular endothelial cells by inhibiting JNK/NF–κB pathways. Sci. Rep. 2015;5(1):1–12. doi: 10.1038/srep08209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liao J.K., Laufs U. Pleiotropic effects of statins. Annu. Rev. Pharmacol. Toxicol. 2005;45:89–118. doi: 10.1146/annurev.pharmtox.45.120403.095748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ahmadi M., et al. Pleiotropic effects of statins: a focus on cancer. Biochim. Biophys. Acta (BBA) - Mol. Basis Dis. 2020;1866(12) doi: 10.1016/j.bbadis.2020.165968. [DOI] [PubMed] [Google Scholar]
  • 73.Emami A., et al. Mechanisms of simvastatin myotoxicity: the role of autophagy flux inhibition. Eur. J. Pharmacol. 2019;862:172616. doi: 10.1016/j.ejphar.2019.172616. [DOI] [PubMed] [Google Scholar]
  • 74.Gao S., et al. Atorvastatin activates autophagy and promotes neurological function recovery after spinal cord injury. Neural Regen. Res. 2016;11(6):977. doi: 10.4103/1673-5374.184498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang J., et al. Statins, autophagy and cancer metastasis. Int. J. Biochem. Cell Biol. 2013;45(3):745–752. doi: 10.1016/j.biocel.2012.11.001. [DOI] [PubMed] [Google Scholar]
  • 76.Peymani P., et al. Statins in patients with COVID-19: a retrospective cohort study in Iranian COVID-19 patients. Transl. Med. Commun. 2021;6(1):1–14. doi: 10.1186/s41231-021-00082-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Prentice E., et al. Coronavirus replication complex formation utilizes components of cellular autophagy. J. Biol. Chem. 2004;279(11):10136–10141. doi: 10.1074/jbc.M306124200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Prentice E., et al. Identification and characterization of severe acute respiratory syndrome coronavirus replicase proteins. J. Virol. 2004;78(18):9977–9986. doi: 10.1128/JVI.78.18.9977-9986.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cottam E.M., et al. Coronavirus nsp6 proteins generate autophagosomes from the endoplasmic reticulum via an omegasome intermediate. Autophagy. 2011;7(11):1335–1347. doi: 10.4161/auto.7.11.16642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chen X., et al. Coronavirus membrane-associated papain-like proteases induce autophagy through interacting with Beclin1 to negatively regulate antiviral innate immunity. Protein Cell. 2014;5(12):912–927. doi: 10.1007/s13238-014-0104-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cottam E.M., Whelband M.C., Wileman T. Coronavirus NSP6 restricts autophagosome expansion. Autophagy. 2014;10(8):1426–1441. doi: 10.4161/auto.29309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Chan J.F.-W., et al. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg. Microb. Infect. 2020;9(1):221–236. doi: 10.1080/22221751.2020.1719902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhao Z., et al. Coronavirus replication does not require the autophagy gene ATG5. Autophagy. 2007;3(6):581–585. doi: 10.4161/auto.4782. [DOI] [PubMed] [Google Scholar]
  • 84.Snijder E.J., et al. Ultrastructure and origin of membrane vesicles associated with the severe acute respiratory syndrome coronavirus replication complex. J. Virol. 2006;80(12):5927–5940. doi: 10.1128/JVI.02501-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Thomas S.A., et al. Countervailing, time-dependent effects on host autophagy promote intracellular survival of Leishmania. J. Biol. Chem. 2018;293(7):2617–2630. doi: 10.1074/jbc.M117.808675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhu E., et al. Classical swine fever virus employs the PERK-and IRE1-dependent autophagy for viral replication in cultured cells. Virulence. 2021;12(1):130–149. doi: 10.1080/21505594.2020.1845040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hawman D.W., et al. Immunocompetent mouse model for Crimean-Congo hemorrhagic fever virus. Elife. 2021;10 doi: 10.7554/eLife.63906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kong W., et al. Mechanisms of mTOR and autophagy in human endothelial cell infected with dengue Virus-2. Viral Immunol. 2020;33(1):61–70. doi: 10.1089/vim.2019.0009. [DOI] [PubMed] [Google Scholar]
  • 89.Campbell G.R., Spector S.A. Vitamin D inhibits human immunodeficiency virus type 1 and Mycobacterium tuberculosis infection in macrophages through the induction of autophagy. PLoS Pathog. 2012;8(5) doi: 10.1371/journal.ppat.1002689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Castro-Gonzalez S., et al. HIV-1 Nef counteracts autophagy restriction by enhancing the association between BECN1 and its inhibitor BCL2 in a PRKN-dependent manner. Autophagy. 2021;17(2):553–577. doi: 10.1080/15548627.2020.1725401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dash S., et al. Hepatitis C virus infection induces autophagy as a prosurvival mechanism to alleviate hepatic ER-stress response. Viruses. 2016;8(5):150. doi: 10.3390/v8050150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Fan X., et al. Foot-and-mouth disease virus infection suppresses autophagy and NF-к B antiviral responses via degradation of ATG5-ATG12 by 3C pro. Cell Death Dis. 2018;8(1) doi: 10.1038/cddis.2016.489. e2561-e2561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Surviladze Z., et al. Cellular entry of human papillomavirus type 16 involves activation of the phosphatidylinositol 3-kinase/Akt/mTOR pathway and inhibition of autophagy. J. Virol. 2013;87(5):2508–2517. doi: 10.1128/JVI.02319-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wang Y., et al. Non‐canonical autophagy functions of ATG16L1 in epithelial cells limit lethal infection by influenza A virus. EMBO J. 2021;40(6) doi: 10.15252/embj.2020105543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yeganeh B., et al. Autophagy activation is required for influenza A virus-induced apoptosis and replication. Biochim. Biophys. Acta Mol. Cell Res. 2018;1865(2):364–378. doi: 10.1016/j.bbamcr.2017.10.014. [DOI] [PubMed] [Google Scholar]
  • 96.Nakamoto M., et al. Virus recognition by Toll-7 activates antiviral autophagy in Drosophila. Immunity. 2012;36(4):658–667. doi: 10.1016/j.immuni.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Han B., Wang Y., Zheng M. Inhibition of autophagy promotes human RSV NS1-induced inflammation and apoptosis in vitro. Exp. Ther. Med. 2021;22(4):1–9. doi: 10.3892/etm.2021.10488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.McLean J.E., et al. Flavivirus NS4A-induced autophagy protects cells against death and enhances virus replication. J. Biol. Chem. 2011;286(25):22147–22159. doi: 10.1074/jbc.M110.192500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.English L., et al. Autophagy enhances the presentation of endogenous viral antigens on MHC class I molecules during HSV-1 infection. Nat. Immunol. 2009;10(5):480–487. doi: 10.1038/ni.1720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Subramanian G., et al. A new mechanism of interferon's antiviral action: induction of autophagy, essential for paramyxovirus replication, is inhibited by the interferon stimulated gene, TDRD7. PLoS Pathog. 2018;14(1) doi: 10.1371/journal.ppat.1006877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lu W., et al. Severe acute respiratory syndrome-associated coronavirus 3a protein forms an ion channel and modulates virus release. Proc. Natl. Acad. Sci. Unit. States Am. 2006;103(33):12540–12545. doi: 10.1073/pnas.0605402103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.He C., Klionsky D.J. Regulation mechanisms and signaling pathways of autophagy. Annu. Rev. Genet. 2009;43:67–93. doi: 10.1146/annurev-genet-102808-114910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wani W.Y., et al. Regulation of autophagy by protein post-translational modification. Lab. Invest. 2015;95(1):14–25. doi: 10.1038/labinvest.2014.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.King K.E., Losier T.T., Russell R.C. Regulation of autophagy enzymes by nutrient signaling. Trends Biochem. Sci. 2021;46(8):687–700. doi: 10.1016/j.tibs.2021.01.006. [DOI] [PubMed] [Google Scholar]
  • 105.Tabibzadeh S. Signaling pathways and effectors of aging. Growth. 2021;3:53. doi: 10.2741/4889. [DOI] [PubMed] [Google Scholar]
  • 106.Zhou Y., et al. Insulin-like growth factor binding protein-related protein 1 activates primary hepatic stellate cells via autophagy regulated by the PI3K/Akt/mTOR signaling pathway. Dig. Dis. Sci. 2020;65(2):509–523. doi: 10.1007/s10620-019-05798-x. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 107.Wang B., et al. High glucose suppresses autophagy through the AMPK pathway while it induces autophagy via oxidative stress in chondrocytes. Cell Death Dis. 2021;12(6):1–13. doi: 10.1038/s41419-021-03791-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bhardwaj M., et al. Seminars in Cancer Biology. Elsevier; 2020. Regulation of autophagy by canonical and non-canonical ER stress responses. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Filomeni G., De Zio D., Cecconi F. Oxidative stress and autophagy: the clash between damage and metabolic needs. Cell Death Differ. 2015;22(3):377–388. doi: 10.1038/cdd.2014.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fang Y., Tan J., Zhang Q. Signaling pathways and mechanisms of hypoxia‐induced autophagy in the animal cells. Cell Biol. Int. 2015;39(8):891–898. doi: 10.1002/cbin.10463. [DOI] [PubMed] [Google Scholar]
  • 111.Juhász G., et al. Gene expression profiling identifies FKBP39 as an inhibitor of autophagy in larval Drosophila fat body. Cell Death Differ. 2007;14(6):1181–1190. doi: 10.1038/sj.cdd.4402123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Son S.M., et al. Autophagy regulation by acetylation—implications for neurodegenerative diseases. Exp. Mol. Med. 2021;53(1):30–41. doi: 10.1038/s12276-021-00556-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Licheva M., et al. Phosphoregulation of the autophagy machinery by kinases and phosphatases. Autophagy. 2021:1–20. doi: 10.1080/15548627.2021.1909407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Jeon M., et al. Regulation of autophagy by protein methylation and acetylation in cancer. J. Cell. Physiol. 2021;237(1):13–28. doi: 10.1002/jcp.30502. [DOI] [PubMed] [Google Scholar]
  • 115.Baek S.H., Kim K.I. Epigenetic control of autophagy: nuclear events gain more attention. Mol. Cell. 2017;65(5):781–785. doi: 10.1016/j.molcel.2016.12.027. [DOI] [PubMed] [Google Scholar]
  • 116.Hu L.-F. Epigenetic regulation of autophagy. Autophagy: Biol. Dis. 2019:221–236. doi: 10.1007/978-981-15-0602-4_11. [DOI] [PubMed] [Google Scholar]
  • 117.Zhu Y., et al. DNA methylation-mediated klotho silencing is an independent prognostic biomarker of head and neck squamous carcinoma. Cancer Manag. Res. 2019;11:1383. doi: 10.2147/CMAR.S188415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Ng K.-M., et al. Amelioration of X-linked related autophagy failure in Danon disease with DNA methylation inhibitor. Circulation. 2016;134(18):1373–1389. doi: 10.1161/CIRCULATIONAHA.115.019847. [DOI] [PubMed] [Google Scholar]
  • 119.Wong J.J.L., Hawkins N.J., Ward R.L. Colorectal cancer: a model for epigenetic tumorigenesis. Gut. 2007;56(1):140–148. doi: 10.1136/gut.2005.088799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Talebian S., et al. The role of epigenetics and non-coding RNAs in autophagy: a new perspective for thorough understanding. Mech. Ageing Dev. 2020 doi: 10.1016/j.mad.2020.111309. [DOI] [PubMed] [Google Scholar]
  • 121.Khalil H., et al. Aging is associated with hypermethylation of autophagy genes in macrophages. Epigenetics. 2016;11(5):381–388. doi: 10.1080/15592294.2016.1144007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Hassen D., et al. Epigenetics reprogramming of autophagy is involved in childhood acute lymphatic Leukemi. Pediatr. Infect. Dis. 2017;2:2573. -282.10045. [Google Scholar]
  • 123.Bai H., et al. A transcriptional variant of the LC3A gene is involved in autophagy and frequently inactivated in human cancers. Oncogene. 2012;31(40):4397–4408. doi: 10.1038/onc.2011.613. [DOI] [PubMed] [Google Scholar]
  • 124.Hu X., et al. Protocadherin 17 acts as a tumour suppressor inducing tumour cell apoptosis and autophagy, and is frequently methylated in gastric and colorectal cancers. J. Pathol. 2013;229(1):62–73. doi: 10.1002/path.4093. [DOI] [PubMed] [Google Scholar]
  • 125.Xie B., et al. Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int. 2013;13(1):1–10. doi: 10.1186/1475-2867-13-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Muhammad J.S., Eladl M.A., Khoder G. Helicobacter pylori-induced DNA methylation as an epigenetic modulator of gastric cancer: recent outcomes and future direction. Pathogens. 2019;8(1):23. doi: 10.3390/pathogens8010023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Tanaka S., et al. Autophagy‐related genes in Helicobacter pylori infection. Helicobacter. 2017;22(3) doi: 10.1111/hel.12376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Muhammad J.S., et al. Autophagy impairment by H elicobacter pylori‐induced methylation silencing of MAP1LC3Av1 promotes gastric carcinogenesis. Int. J. Cancer. 2017;140(10):2272–2283. doi: 10.1002/ijc.30657. [DOI] [PubMed] [Google Scholar]
  • 129.Sengupta S., et al. Mycobacterium tuberculosis phosphoribosyltransferase promotes bacterial survival in macrophages by inducing histone hypermethylation in autophagy-related genes. Front. Cell. Infect. Microbiol. 2021:11. doi: 10.3389/fcimb.2021.676456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Hu L., et al. Crystal structure of TET2-DNA complex: insight into TET-mediated 5mC oxidation. Cell. 2013;155(7):1545–1555. doi: 10.1016/j.cell.2013.11.020. [DOI] [PubMed] [Google Scholar]
  • 131.Fu R., et al. TET1 exerts its tumour suppressor function by regulating autophagy in glioma cells. Biosci. Rep. 2017;37(6) doi: 10.1042/BSR20160523. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 132.Fu R., et al. Ten-eleven translocation 1 regulates methylation of autophagy-related genes in human glioma. Neuroreport. 2018;29(9):731–738. doi: 10.1097/WNR.0000000000001024. [DOI] [PubMed] [Google Scholar]
  • 133.Peng J., et al. Tet methylcytosine dioxygenase 2 inhibits atherosclerosis via upregulation of autophagy in ApoE−/− mice. Oncotarget. 2016;7(47):76423. doi: 10.18632/oncotarget.13121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Bánréti Á., Sass M., Graba Y. The emerging role of acetylation in the regulation of autophagy. Autophagy. 2013;9(6):819–829. doi: 10.4161/auto.23908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Füllgrabe J., et al. The histone H4 lysine 16 acetyltransferase hMOF regulates the outcome of autophagy. Nature. 2013;500(7463):468–471. doi: 10.1038/nature12313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Füllgrabe J., et al. Cracking the survival code: autophagy-related histone modifications. Autophagy. 2014;10(4):556–561. doi: 10.4161/auto.27280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Huang R., et al. Deacetylation of nuclear LC3 drives autophagy initiation under starvation. Mol. Cell. 2015;57(3):456–466. doi: 10.1016/j.molcel.2014.12.013. [DOI] [PubMed] [Google Scholar]
  • 138.Nardacci R., et al. Role of autophagy in HIV infection and pathogenesis. J. Intern. Med. 2017;281(5):422–432. doi: 10.1111/joim.12596. [DOI] [PubMed] [Google Scholar]
  • 139.Demonté D., et al. Administration of HDAC inhibitors to reactivate HIV-1 expression in latent cellular reservoirs: implications for the development of therapeutic strategies. Biochem. Pharmacol. 2004;68(6):1231–1238. doi: 10.1016/j.bcp.2004.05.040. [DOI] [PubMed] [Google Scholar]
  • 140.Moreira J.D., et al. Functional inhibition of host histone deacetylases (HDACs) enhances in vitro and in vivo anti-mycobacterial activity in human macrophages and in zebrafish. Front. Immunol. 2020;11:36. doi: 10.3389/fimmu.2020.00036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Marín N.D., et al. Functional profile of CD4+ and CD8+ T cells in latently infected individuals and patients with active TB. Tuberculosis. 2013;93(2):155–166. doi: 10.1016/j.tube.2012.12.002. [DOI] [PubMed] [Google Scholar]
  • 142.Fu S., et al. Crosstalk between hepatitis B virus X and high‐mobility group box 1 facilitates autophagy in hepatocytes. Mol. Oncol. 2018;12(3):322–338. doi: 10.1002/1878-0261.12165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Mercer T.R., Dinger M.E., Mattick J.S. Long non-coding RNAs: insights into functions. Nat. Rev. Genet. 2009;10(3):155–159. doi: 10.1038/nrg2521. [DOI] [PubMed] [Google Scholar]
  • 144.Zhao G., et al. The long noncoding RNA MALAT 1 regulates the lipopolysaccharide‐induced inflammatory response through its interaction with NF‐κB. FEBS (Fed. Eur. Biochem. Soc.) Lett. 2016;590(17):2884–2895. doi: 10.1002/1873-3468.12315. [DOI] [PubMed] [Google Scholar]
  • 145.Cech T.R., Steitz J.A. The noncoding RNA revolution—trashing old rules to forge new ones. Cell. 2014;157(1):77–94. doi: 10.1016/j.cell.2014.03.008. [DOI] [PubMed] [Google Scholar]
  • 146.Choudhry H., Harris A.L., McIntyre A. The tumour hypoxia induced non-coding transcriptome. Mol. Aspect. Med. 2016;47:35–53. doi: 10.1016/j.mam.2016.01.003. [DOI] [PubMed] [Google Scholar]
  • 147.Smith-Vikos T., Slack F.J. MicroRNAs and their roles in aging. J. Cell Sci. 2012;125(1):7–17. doi: 10.1242/jcs.099200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Gozuacik D., et al. Autophagy-regulating microRNAs and cancer. Front. Oncol. 2017;7:65. doi: 10.3389/fonc.2017.00065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wang Y., Wang Q., Song J. Inhibition of autophagy potentiates the proliferation inhibition activity of microRNA-7 in human hepatocellular carcinoma cells. Oncol. Lett. 2017;14(3):3566–3572. doi: 10.3892/ol.2017.6573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Xu Y., et al. miR-101 inhibits autophagy and enhances cisplatin-induced apoptosis in hepatocellular carcinoma cells. Oncol. Rep. 2013;29(5):2019–2024. doi: 10.3892/or.2013.2338. [DOI] [PubMed] [Google Scholar]
  • 151.Valera E., et al. MicroRNA-101 modulates autophagy and oligodendroglial alpha-synuclein accumulation in multiple system atrophy. Front. Mol. Neurosci. 2017;10:329. doi: 10.3389/fnmol.2017.00329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Wang Z., et al. Ultra bright red AIE dots for cytoplasm and nuclear imaging. Polym. Chem. 2014;5(24):7013–7020. [Google Scholar]
  • 153.Silwal P., et al. Seminars in Cell & Developmental Biology. Elsevier; 2020. The roles of microRNAs in regulation of autophagy during bacterial infection. [DOI] [PubMed] [Google Scholar]
  • 154.Kumar R., et al. MicroRNA 17‐5p regulates autophagy in Mycobacterium tuberculosis‐infected macrophages by targeting Mcl‐1 and STAT3. Cell Microbiol. 2016;18(5):679–691. doi: 10.1111/cmi.12540. [DOI] [PubMed] [Google Scholar]
  • 155.Duan X., et al. microRNA-17-5p modulates bacille calmette-guerin growth in RAW264. 7 cells by targeting ULK1. PLoS One. 2015;10(9) doi: 10.1371/journal.pone.0138011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pires D., et al. Mycobacterium tuberculosis modulates miR-106b-5p to control cathepsin S expression resulting in higher pathogen survival and poor T-cell activation. Front. Immunol. 2017;8:1819. doi: 10.3389/fimmu.2017.01819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Lu C., et al. MIR106B and MIR93 prevent removal of bacteria from epithelial cells by disrupting ATG16L1-mediated autophagy. Gastroenterology. 2014;146(1):188–199. doi: 10.1053/j.gastro.2013.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Yang L., et al. Long non-coding RNAs involved in autophagy regulation. Cell Death Dis. 2017;8(10) doi: 10.1038/cddis.2017.464. e3073-e3073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Gupta R.A., et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464(7291):1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Ying L., et al. Downregulated MEG3 activates autophagy and increases cell proliferation in bladder cancer. Mol. Biosyst. 2013;9(3):407–411. doi: 10.1039/c2mb25386k. [DOI] [PubMed] [Google Scholar]
  • 161.Pawar K., et al. Down regulated lncRNA MEG3 eliminates mycobacteria in macrophages via autophagy. Sci. Rep. 2016;6(1):1–13. doi: 10.1038/srep19416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Xiong H., et al. LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene. 2017;36(25):3528–3540. doi: 10.1038/onc.2016.521. [DOI] [PubMed] [Google Scholar]
  • 163.Enuka Y., et al. Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res. 2016;44(3):1370–1383. doi: 10.1093/nar/gkv1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Han B., Chao J., Yao H. Circular RNA and its mechanisms in disease: from the bench to the clinic. Pharmacol. Ther. 2018;187:31–44. doi: 10.1016/j.pharmthera.2018.01.010. [DOI] [PubMed] [Google Scholar]
  • 165.Verduci L., et al. CircRNAs: role in human diseases and potential use as biomarkers. Cell Death Dis. 2021;12(5):1–12. doi: 10.1038/s41419-021-03743-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Wang Y., et al. The influence of circular RNAs on autophagy and disease progression. Autophagy. 2021:1–14. doi: 10.1080/15548627.2021.1917131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Yao L., Xu B., Li X. Neisseria gonorrhoeae-induced salpingitis is targeted by circular RNA EIF3K via miR-139-5p and regulating MAPK/NF-κB signaling pathway to promotes apoptosis and autophagy bacterial cells. Microb. Pathog. 2020;142 doi: 10.1016/j.micpath.2020.104051. [DOI] [PubMed] [Google Scholar]
  • 168.Zheng J., et al. CircCDK8 regulates osteogenic differentiation and apoptosis of PDLSCs by inducing ER stress/autophagy during hypoxia. Ann. N. Y. Acad. Sci. 2021;1485(1):56–70. doi: 10.1111/nyas.14483. [DOI] [PubMed] [Google Scholar]
  • 169.Zhu M., et al. Exosomes derived from mmu_circ_0000623-modified ADSCs prevent liver fibrosis via activating autophagy. Hum. Exp. Toxicol. 2020;39(12):1619–1627. doi: 10.1177/0960327120931152. [DOI] [PubMed] [Google Scholar]
  • 170.Cheng Y., et al. CircRNA-012091/PPP1R13B–mediated lung fibrotic response in silicosis via endoplasmic reticulum stress and autophagy. Am. J. Respir. Cell Mol. Biol. 2019;61(3):380–391. doi: 10.1165/rcmb.2019-0017OC. [DOI] [PubMed] [Google Scholar]
  • 171.Li X.-X., et al. Interaction between HuR and circPABPN1 modulates autophagy in the intestinal epithelium by altering ATG16L1 translation. Mol. Cell. Biol. 2020;40(6):e00492–19. doi: 10.1128/MCB.00492-19. e00492-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Yu T., et al. Circular RNA GATAD2A promotes H1N1 replication through inhibiting autophagy. Vet. Microbiol. 2019;231:238–245. doi: 10.1016/j.vetmic.2019.03.012. [DOI] [PubMed] [Google Scholar]
  • 173.Fang Y., et al. MicroRNA‐7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3‐kinase/Akt pathway in hepatocellular carcinoma. Hepatology. 2012;55(6):1852–1862. doi: 10.1002/hep.25576. [DOI] [PubMed] [Google Scholar]
  • 174.Frankel L.B., et al. microRNA‐101 is a potent inhibitor of autophagy. EMBO J. 2011;30(22):4628–4641. doi: 10.1038/emboj.2011.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Li S., et al. MiR-20a and miR-20b negatively regulate autophagy by targeting RB1CC1/FIP200 in breast cancer cells. Life Sci. 2016;147:143–152. doi: 10.1016/j.lfs.2016.01.044. [DOI] [PubMed] [Google Scholar]
  • 176.Wang Z., et al. MicroRNA-25 regulates chemoresistance-associated autophagy in breast cancer cells, a process modulated by the natural autophagy inducer isoliquiritigenin. Oncotarget. 2014;5(16):7013. doi: 10.18632/oncotarget.2192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Zhai Z., et al. miR-106b fine tunes ATG16L1 expression and autophagic activity in intestinal epithelial HCT116 cells. Inflamm. Bowel Dis. 2013;19(11):2295–2301. doi: 10.1097/MIB.0b013e31829e71cf. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Xu N., et al. Cisplatin-induced downregulation of miR-199a-5p increases drug resistance by activating autophagy in HCC cell. Biochem. Biophys. Res. Commun. 2012;423(4):826–831. doi: 10.1016/j.bbrc.2012.06.048. [DOI] [PubMed] [Google Scholar]
  • 179.Chatterjee A., Chattopadhyay D., Chakrabarti G. miR-17-5p downregulation contributes to paclitaxel resistance of lung cancer cells through altering beclin1 expression. PLoS One. 2014;9(4):e95716. doi: 10.1371/journal.pone.0095716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Chen Y., et al. Identification of ULK1 as a novel biomarker involved in miR-4487 and miR-595 regulation in neuroblastoma SH-SY5Y cell autophagy. Sci. Rep. 2015;5(1):1–10. doi: 10.1038/srep11035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Tan S., et al. miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int. J. Mol. Med. 2016;37(4):1030–1038. doi: 10.3892/ijmm.2016.2492. [DOI] [PubMed] [Google Scholar]
  • 182.Stresemann C., Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int. J. Cancer. 2008;123(1):8–13. doi: 10.1002/ijc.23607. [DOI] [PubMed] [Google Scholar]
  • 183.Karahoca M., Momparler R.L. Pharmacokinetic and pharmacodynamic analysis of 5-aza-2’-deoxycytidine (decitabine) in the design of its dose-schedule for cancer therapy. Clin. Epigenet. 2013;5(1):1–16. doi: 10.1186/1868-7083-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Foggetti G., et al. Autophagy induced by SAHA affects mutant P53 degradation and cancer cell survival. Biosci. Rep. 2019;39(2) doi: 10.1042/BSR20181345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Shao Y., et al. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc. Natl. Acad. Sci. Unit. States Am. 2004;101(52):18030–18035. doi: 10.1073/pnas.0408345102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.New M., et al. A regulatory circuit that involves HR23B and HDAC6 governs the biological response to HDAC inhibitors. Cell Death Differ. 2013;20(10):1306–1316. doi: 10.1038/cdd.2013.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lee J.Y.-C., et al. Inhibition of HDAC3-and HDAC6-promoted survivin expression plays an important role in SAHA-induced autophagy and viability reduction in breast cancer cells. Front. Pharmacol. 2016;7:81. doi: 10.3389/fphar.2016.00081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Watanabe M., et al. Induction of autophagy in malignant rhabdoid tumor cells by the histone deacetylase inhibitor FK228 through AIF translocation. Int. J. Cancer. 2009;124(1):55–67. doi: 10.1002/ijc.23897. [DOI] [PubMed] [Google Scholar]
  • 189.Hui K.F., Yeung P.L., Chiang A.K. Induction of MAPK-and ROS-dependent autophagy and apoptosis in gastric carcinoma by combination of romidepsin and bortezomib. Oncotarget. 2016;7(4):4454. doi: 10.18632/oncotarget.6601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Mehrpouri M., et al. Anti-leukemic effects of histone deacetylase (HDAC) inhibition in acute lymphoblastic leukemia (ALL) cells: shedding light on mitigating effects of NF-κB and autophagy on panobinostat cytotoxicity. Eur. J. Pharmacol. 2020;875 doi: 10.1016/j.ejphar.2020.173050. [DOI] [PubMed] [Google Scholar]
  • 191.El Baba R., Herbein G. Management of epigenomic networks entailed in coronavirus infections and COVID-19. Clin. Epigenet. 2020;12(1):1–12. doi: 10.1186/s13148-020-00912-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Koyuncu E., et al. Sirtuins are evolutionarily conserved viral restriction factors. mBio. 2014;5(6) doi: 10.1128/mBio.02249-14. e02249-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Gordon D.E., et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. 2020;583(7816):459–468. doi: 10.1038/s41586-020-2286-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Ruiz‐Magaña M.J., et al. The DNA methyltransferase inhibitors zebularine and decitabine induce mitochondria‐mediated apoptosis and DNA damage in p53 mutant leukemic T cells. Int. J. Cancer. 2012;130(5):1195–1207. doi: 10.1002/ijc.26107. [DOI] [PubMed] [Google Scholar]
  • 195.Kong Y.-L., et al. Chidamide, a histone deacetylase inhibitor, inhibits autophagy and exhibits therapeutic implication in chronic lymphocytic leukemia. Aging (Albany NY) 2020;12(16):16083. doi: 10.18632/aging.103536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Angeletti F., et al. Inhibition of the autophagy pathway synergistically potentiates the cytotoxic activity of givinostat (ITF2357) on human glioblastoma cancer stem cells. Front. Mol. Neurosci. 2016;9:107. doi: 10.3389/fnmol.2016.00107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.El-Khoury V., et al. Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia. 2014;28(8):1636–1646. doi: 10.1038/leu.2014.19. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data will be made available on request.


Articles from Biochemistry and Biophysics Reports are provided here courtesy of Elsevier

RESOURCES