Next Article in Journal
Enhancement of the Colorectal Chemopreventive and Immunization Potential of Northern Thai Purple Rice Anthocyanin Using the Biotransformation by β-Glucosidase-Producing Lactobacillus
Next Article in Special Issue
Hydrogen Sulfide: A Key Role in Autophagy Regulation from Plants to Mammalians
Previous Article in Journal
Identification of Regulatory Factors and Prognostic Markers in Amyotrophic Lateral Sclerosis
Previous Article in Special Issue
Conserved and Diversified Mechanism of Autophagy between Plants and Animals upon Various Stresses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How Cells Deal with the Fluctuating Environment: Autophagy Regulation under Stress in Yeast and Mammalian Systems

1
Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
2
Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2022, 11(2), 304; https://doi.org/10.3390/antiox11020304
Submission received: 2 January 2022 / Revised: 28 January 2022 / Accepted: 31 January 2022 / Published: 2 February 2022
(This article belongs to the Special Issue Regulation of Autophagy under Stress)

Abstract

:
Eukaryotic cells frequently experience fluctuations of the external and internal environments, such as changes in nutrient, energy and oxygen sources, and protein folding status, which, after reaching a particular threshold, become a type of stress. Cells develop several ways to deal with these various types of stress to maintain homeostasis and survival. Among the cellular survival mechanisms, autophagy is one of the most critical ways to mediate metabolic adaptation and clearance of damaged organelles. Autophagy is maintained at a basal level under normal growing conditions and gets stimulated by stress through different but connected mechanisms. In this review, we summarize the advances in understanding the autophagy regulation mechanisms under multiple types of stress including nutrient, energy, oxidative, and ER stress in both yeast and mammalian systems.

1. Overview of Autophagy in Yeast and Mammals

Autophagy is a highly regulated cellular degradation and recycling process, conserved from yeast to more complex eukaryotes [1]. The proteasome is responsible for degrading most short-lived, individual proteins, therefore, autophagy can degrade and recycle long-lived proteins, large protein complexes, and organelles [2]. The key definition of autophagy is the delivery and, typically, the degradation of cytoplasmic cargo within the lysosome (or the vacuole in fungi and plants) [3]. Based on different types of cargo and various modes of cargo delivery, at least three types of autophagy have been characterized, including microautophagy, macroautophagy, and chaperone-mediated auto-phagy/CMA; the latter process occurs in birds, fish, and mammals, but is not present in fungi [4]. The most comprehensively studied of these processes is macroautophagy, and, hereafter, we will use the term autophagy to refer to macroautophagy. The morphological hallmark of autophagy is the formation of the autophagosome, a large cytoplasmic double-membrane vesicle, which originates through the generation of the phagophore and the latter’s subsequent expansion and closure [5]. Once completed, the outer membrane of the autophagosome fuses with the lysosome/vacuole, while the inner membrane and cargo are exposed to the lumen of the degradative organelles for hydrolysis and the final efflux of the breakdown products into the cytosol [6].
The basic mechanism of autophagy has been well-documented, and the entire process of autophagy can be divided into the following stages: induction and nucleation of the phagophore, expansion and maturation of the phagophore into a completed auto-phagosome, docking and fusion with the lysosome/vacuole, and degradation and efflux of the breakdown products (Figure 1A) [7]. Initially identified in yeast, over 40 genes that have products primarily involved in the basic process of autophagy have been classified under the name autophagy-related (ATG) [8]. Many papers have provided details on this topic [2,4,9], therefore, here, we will briefly describe the autophagy process and the Atg proteins involved in both yeast and mammalian systems.
In yeast, the induction of autophagy begins at a single perivacuolar site, called the phagophore assembly site (PAS) which is proximal to the vacuole. This step is regulated by the Atg1 protein complex, including Atg1, Atg13, and the Atg17-Atg31-Atg29 ternary subcomplex [10,11]. In the nucleation stage, the Atg14-containing class III phosphatidylinositol (PtdIns) 3-kinase (PtdIns3K) complex I (consisting of Vps34, Vps30, Vps15, Atg14, and Atg38) is recruited to the PAS (Figure 1B) [12]. Next, the phagophore begins to expand and then seal to complete the formation of the autophagosome. Key components participating at this stage are two ubiquitin-like (Ubl) conjugation systems, which mediate the conjugation of Ubl proteins Atg12 and Atg8 [13,14] (Figure 1C). Through an enzymatic pathway involving Atg7 (an E1-like enzyme) for Atg12 activation and Atg10 (an E2-like enzyme) for Atg12–Atg5 conjugation, the C terminus of Atg12 is conjugated to an internal Lys of Atg5; Atg16 then noncovalently binds to Atg5 in the conjugate [15]. This system plays a role in membrane recruitment for the expanding phagophore. In contrast to Atg12, which is conjugated to another protein, Atg8 is conjugated to the lipid phosphatidylethanolamine (PE), allowing for its membrane association. Atg8 is initially synthesized with a C-terminal extension, which is removed by the Atg4 cysteine protease to expose a C-terminal Gly [16]. The modified Atg8 is activated with the help of Atg7, and then transferred to Atg3 (an E2 enzyme) that attaches the exposed C-terminal Gly to PE [13,15]. Atg8–PE is found on both sides of the phagophore and initially of the autophagosome; the portion on the autophagosome outer membrane will be deconjugated by a second Atg4-depedent cleavage when autophagosome formation is completed. The transmembrane protein Atg9 may cycle between the PAS and peripheral sites, thus carrying or directing the delivery of membrane for the expansion stage [17]. Upon maturation, the intact autophagosome fully surrounds the cargo, and ultimately delivers cargos to the vacuole by fusing with the vacuolar membrane. Finally, the cargo is degraded by various hydrolases in the vacuole, and breakdown products will be released back into the cytoplasm through permeases in the vacuole membrane.
There are some slight differences in components involved in the autophagy process in mammalian cells, whereas most components are homologs of Atg proteins in yeast. The activation of the ULK Ser/Thr kinase complex is required for autophagy induction, and the initiation begins with the ULK kinase complex (the catalytic subunits ULK1 or ULK2, the regulatory scaffold protein ATG13, RB1CC1, and the stabilizing protein ATG101) which can phosphorylate downstream factors for the induction of autophagy [9,18]. Next, the activated ULK1 complex phosphorylates and activates the PtdIns3K complex 1 (mainly composed of BECN1 [beclin 1], PIK3C3/VPS34, PIK3R4/VPS15, ATG14, NRBF2, and AMBRA1) [19]. The activated PIK3C3/VPS34 can phosphorylate PtdIns to produce phosphatidylinositol-3-phosphate (PtdIns3P), further contributing to formation of the phagophore [20]; ATG14 is directly associated with the ability of the PtdIns3K complex I to translocate to this site [21]. NRBF2 regulates PIK3C3 activity via promoting assembly of the complex, while AMBRA1 contributes to the interaction of BECN1 and PIK3C3 and the catalytic activity [22,23] (Figure 1B). Phagophores are nucleated on ER-emanating PtdIns3P-rich membrane domains called omegasomes [24]. In the expansion step, the ATG12 conjugation system (ATG12–ATG5-ATG16L1 complex) is like that in yeast (Figure 1C). In addition, the second Ubl system involves Atg8-family proteins including MAP1LC3/LC3 and GABARAP subfamilies, undergoing a similar process. With another protein, UVRAG, and core proteins from the PtdIns3K complex I, PtdIns3K complex II is formed, which is also important for autophagy. For example, UVRAG can bind to SH3GLB1 and promote autophagosome maturation [19].
Under normal conditions, autophagy keeps working constitutively at a basal state to maintain cellular homeostasis. When the cell is exposed to certain stress conditions, autophagy is massively induced and promotes the turnover of cytoplasmic materials required for cell survival or removing superfluous or damaged organelles. Too little or too much degradation from uncontrolled autophagy is harmful, and aberrant autophagy is associated with various diseases, such as cancer, aging, and neurodegeneration [25]. Autophagy can be either nonselective or selective: the nonselective mode degrades relatively random portions of the cytoplasm (although phase separation may be involved), whereas the selective mode is highly specific for certain components [26]. In particular, selective autophagy can degrade damaged and superfluous organelles or invasive microbes; these selective processes are given different names depending on the cargo, including mito-phagy (mitochondria), pexophagy (peroxisomes), aggrephagy (protein aggregates), lipophagy (lipid droplets), and xenophagy (intracellular pathogens) [26]. The selective mode of autophagy also plays a key role in cell physiology.
The frequently changing external environment causes cellular stress, and cells in a diseased state also experience stress from the unstable internal environment; therefore, stress biomarkers and their detection are important in the assessment of cell homeostasis (Table 1). To meditate metabolic adaptation and clear damaged organelles, autophagy is considered as one of the most important mechanisms to maintain cell survival under stress. Whereas the depletion of nutrients constitutes the main stimulus for massive autophagy induction, many other types of cellular stress are also involved in autophagy regulation. These different conditions can also regulate autophagy at different levels, including epigenetic, transcription, post-transcription, translation, and post-translation. In this review, we consider different stress stimuli and their relations with autophagy, with a goal of providing a more comprehensive understanding about this field.

2. Autophagy Regulation under Nutrient Stress

2.1. Mechanisms of Autophagy Regulation by Nutrient Stress in Yeast

Nutrients, such as amino acids and other nitrogen sources, are crucial for yeast growth and the target of rapamycin (TOR) pathway is the central regulator [52]. Tor1 and Tor2 are conserved protein kinases that can be found in two protein complexes termed TOR complex 1 (TORC1) and TORC2. TORC1, which consists of Tor1 or Tor2, Kog1, Lst8, and Tco89, is particularly sensitive to rapamycin treatment and activated by nutrients [52]. Unlike TORC1, TORC2 can only utilize Tor2 as its catalytic component and is not sensitive to rapamycin [53]. Even though some studies indicate that TORC2 is involved in promoting autophagy [54], TORC1 is still considered as the master regulator of autophagy, especially under nutrient stress.
Nitrogen and amino acid signaling are transmitted to TORC1 via different mechanisms, which largely involve the conserved RAG GTPase, composed of Gtr1 and Gtr2 [55]. Gtr1 and Gtr2 form a heterodimer and are tethered on the vacuole membrane by the Ego1-Ego2-Ego3/EGO complex [52]. In the presence of sufficient nitrogen sources/amino acids, with the help of Vam6 as a guanine nucleotide exchange factor/GEF, Gtr1 and Gtr2 are in the GTP and GDP bound forms, respectively. Activated Gtr1 binds to Kog1 and Tco89 in the TORC1 complex, trapping and activating TORC1 on the vacuole [56]. When the amino acid level goes down, Seh1-associated subcomplex inhibiting TORC1/SEACIT, which is composed of Npr2, Npr3, and the catalytic subunit Iml1, functions as the GTPase activating protein (GAP) to induce GDP loading onto Gtr1; this form no longer activates TORC1 [57].
TORC1 regulation function with regard to autophagy can be summarized in three aspects (Figure 2A). First, TORC1 directly phosphorylates Atg13 under growing conditions, which prevents Atg13-Atg1 complex activity [58,59]. The inactivation of TORC1 by nutrient deprivation leads to the hypophosphorylation of Atg13, the induction of Atg1 kinase activity and autophagy stimulation [58].
Second, TORC1 regulates the transcription of ATG genes. TORC1 negatively regulates the expression of genes required for the adaptation to nutrient stress by regulating the expression and localization of several transcription factors (TFs), including Gcn4, and the GATA-binding proteins Gln3 and Gat1 [60,61]. When TORC1 is inactivated by the depletion of nutrients, the activated phosphatases Sit4 and PP2A (Pph21/22-Tpd3-Cdc55) mediate the translocation of Gln3 and Gat1 to the nucleus [60], where they are required for the successful induction of ATG7, ATG8, ATG9, ATG29, and ATG32 expression [62]; Gln3 is necessary for promoting ATG14 expression [63]. TORC1 inhibition also induces Gcn4 expression [61] and GCN4 deletion leads to decreased ATG1 mRNA level and autophagy activity during starvation [62]. Additionally, Gcn4 is responsible for the increased expression of ATG41 during starvation, which is required for efficient autophagy [64]. In addition to directly regulating the localization of TFs, TORC1 and one of its downstream targets, Sch9, inhibit the translocation of Rim15 from the cytosol to the nucleus; Rim15 is a kinase, which controls the association between several TFs and ATG genes [27,65]. For instance, Rph1 is a transcriptional repressor of several ATG genes, including ATG7, ATG8, ATG9, ATG14, and ATG29 under nutrient-rich conditions. Upon nutrient stress, Rph1 is phosphorylated by the nuclear-localized Rim15 and dissociates from the ATG genes to induce autophagy [66]. Similarly, Ume6 inhibits ATG8 transcription when nutrients are abundant, and this inhibition is relieved by Rim15-dependent phosphorylation during starvation [67].
Third, TORC1 controls the posttranscriptional regulation of ATG genes. In a report by Hu et al., a temperature sensitive mutation in the decapping enzyme Dcp2 results in increasing mRNA level of multiple ATG genes under nutrient-replete conditions, including ATG1, ATG8, ATG9, and ATG13, and a higher autophagy activity [68]. In the same study, the researchers also found that in C. neoformans, TORC1 phosphorylates Dcp2 under nutrient-rich condition, thus promoting ATG8 mRNA degradation [68].
Here, we only summarized autophagy regulation related to TORC1. Multiple layers of regulation happen in both growing and/or starvation conditions to regulate autophagy as listed in Table 2, but their relations to nutrient stress signals are not completely elucidated.
Although additional factors have been identified that regulate autophagy in yeast during starvation, many questions still remain. First, some autophagy regulators play dual roles under growing and starvation conditions (Table 2). For example, Dhh1 contributes to the degradation of multiple ATG mRNAs under nutrient-rich conditions, but, on the contrary, promotes the translation of ATG genes during starvation [68,73,81]. However, how this transition happens and how it connects with a nutrient-sensing pathway is still unclear. Second, TORC1 has different localizations based on nutrient status. When nutrients are replete, TORC1 is activated and disperses along the vacuole membrane; several studies indicate that TORC1 forms punctate structure on the vacuole in response to starvation [82,83,84]. However, it is still unclear whether the change in TORC1 localization contributes to autophagy regulation. One model proposes that because the PAS is formed close to the vacuole, the dispersed TORC1 localization along the vacuole prevents Atg13 recruitment to the PAS. Conversely, TORC1 puncta formation during starvation limits its access to Atg13, providing more opportunities for hypophosphorylated Atg13 to be recruited to the PAS [82]. A recent study indicates that the EGO complex and TORC1 have two localizations, both on endosomes and the vacuole but only the TORC1 on endosomes controls autophagy through targeting Atg13 [85]. However, it is not clear why TORC1 has these two different pools and, considering that the distribution between them does not change during nitrogen starvation, why these two populations of TORC1 function differently. Third, epigenetic regulation is another critical way to control autophagy at an appropriate level. Although not much is known in yeast, some evidence indicates the relationship between TORC1, histone modifications, and autophagy regulation. A study from Füllgrabe et al. identified that autophagy occurs concomitant with the reduction in histone H4 Lys16 acetylation/H4K16ac, which may result from the autophagic degradation of the acetyltransferase Sas2 [80]. More recently, Set2, a histone methyltransferase, was shown to be necessary for the transcriptional response to nutrient stress; Set2 genetically interacts with Tor1 and Tor2, indicating a potential role in autophagy regulation [86], but further studies are needed to reveal the mechanism of this type of regulation.

2.2. Autophagy Regulation in Mammalian Cells

In mammalian cells, nutrient starvation is also a common stress that induces auto-phagy. Multiple important nutrient-response molecules have been reported to regulate autophagy, among which MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) is the best characterized. In this subsection, we will summarize autophagy regulation mechanisms mediated by MTORC1 and briefly introduce some other molecules that contribute to autophagy regulation under nutrient stress.

2.2.1. Autophagy Regulation by MTORC1

Like in yeast, there are two TOR complexes in mammalian cells, MTORC1 and MTORC2. MTORC1, which consists of MTOR, RPTOR/raptor, DEPTOR, LST8, and PRAS40, is the general responder to growth factors and nutrients [87]. Amino acids are essential for the activation of MTORC1 through RRAG GTPases [88,89]. Mammalian cells contain four RRAG GTPase members, RRAGA, RRAGB, RRAGC, and RRAGD and they functions in a heterodimer, in which one monomer of either RRAGA or RRAGB partners with either RRAGC or RRAGD [90]. Amino acids in the lysosomal lumen activate the Ragulator complex, possibly through the vacuolar-type H+-translocating ATPase (V-ATPase) [91,92], and the Ragulator complex functions as a guanine nucleotide exchange factor that promotes the active conformation of RRAG GTPase; where RRAGA/B binds with GTP and RRAGC/D is loaded with GDP [91,92,93]. In addition, amino acids inhibit the GATOR1 complex (analogous to yeast SEACIT), the GAP for RRAGA/B, therefore facilitating the activation of the RRAG GTPase [94]. Once activated, RRAG heterodimer binds with RPTOR and brings MTORC1 into proximity with RHEB (Ras homolog, mTORC1 binding) GTPase on lysosomes [88]. MTORC1 activity is coupled with growth factors; the removal of TSC1-TSC2, the GAP of RHEB GTPase, in response to growth factors, allows the activation of MTORC1 by RHEB [95]. In contrast, the lack of nutrients results in the conversion of RRAG GTPase into its inactive form and the lysosomal localization of TSC2, which inhibits RHEB GTPase activity [96]. In addition, the absence of amino acids inhibits the polyubiquitination of RHEB, which is important for its binding with MTORC1 [97]. As a result, MTORC1 becomes inactivated and displays a cytosolic localization.
MTORC1 is considered as the master regulator of autophagy when cells are facing nutrient stress. Autophagy regulation by MTORC1 can be summarized in the following aspects (Figure 2B): First, MTORC1 regulates the posttranslational modification of autophagy-associated proteins. Several ATG proteins are the direct targets of MTORC1 and the best known are ULK1 and ATG13. MTORC1-dependent phosphorylation of ULK1 and ATG13 reduces ULK1 complex activity. During starvation, the inactivated MTORC1 disassociates from ULK1, relieving inhibition of the latter; subsequent triggering of ULK1 complex activity promotes autophagy [98,99,100]. In return, activated ULK1 inhibits MTORC1 activity via phosphorylating RPTOR and reducing its substrate-binding ability [101,102]. This feedback loop maintains the inactivation of MTORC1 and is important for the full activation of autophagy during nutrient deprivation.
Several components of the PtdIns3K complex are also targets of MTORC1. In complex I, MTORC1-dependent phosphorylation of ATG14 inhibits the kinase activity of the complex [103]. Another component, NRBF2, can be phosphorylated by MTORC1 at Ser113 and Ser120. MTORC1 inhibition suppresses NRBF2 phosphorylation and changes it binding preference from PIK3C3/VPS34 and PIK3R4/VPS15 to ATG14 and BECN1, supporting PtdIns3K complex I assembly and its association with the ULK1 complex [104]. In addition, AMBRA1 is phosphorylated by MTORC1 at Ser52 and becomes inactivated. Upon nutrient stress, activated AMBRA1 interacts with the E3 ligase TRAF6 and ubiquitinates ULK1, enhancing its activity [105]. In complex II, UVRAG is the direct target of MTORC1 [106]. MTORC1-dependent phosphorylation on Ser498 has a positive effect on the interaction between UVRAG and RUBCN, which negatively regulates PIK3C3/VPS34 kinase activity and the interaction between HOPS (a complex involved in tethering) and UVRAG, therefore inhibiting the fusion between autophagosomes and lysosomes [106]. Apart from the proteins in these two complexes, WIPI2 can be phosphorylated by MTORC1 at Ser395, which promotes its polyubiquitination by HUWE1 and subsequent degradation [107].
Besides directly phosphorylating ATG proteins, MTORC1-dependent phosphorylation of the acetyltransferase EP300 prevents its intra-molecular inhibition, thus activating its catalytic activity [108]. Several ATG proteins, including ATG5, ATG7, ATG8, and ATG12 are the targets of EP300, and the acetylation of these ATG proteins inhibits autophagy [109]. Therefore, under nutrient stress, MTORC1 inactivation reduces the acetylation of essential ATG proteins to fully activate autophagy.
Second, MTORC1 controls the transcription of ATG genes and lysosomal genes via regulating the localization of several TFs. TFEB is a member of the basic helix-loop-helix leucine-zipper family of TFs that promotes the transcription of genes in lysosomal biogenesis and autophagy [110]. When nutrients are repleted, TFEB will be recruited to the lysosome by active RRAG GTPase and phosphorylated by MTORC1 [111], whereas starvation leads to a rapid translocation of TFEB from the cytosol to the nucleus and the induction of transcription of autophagy-associated genes such as UVRAG, WIPI, MAPLC3B, SQSTM1, VPS11, VPS18, and ATG9B [112]. Several MTORC1-dependent phosphorylation sites are found on TFEB, including Ser122, Ser138, Ser142, and Ser211 [113,114,115,116], which regulate TFEB cellular localization through different but coordinated mechanisms. Phosphorylation on Ser211 by MTORC1 promotes TFEB association with YWHA/14-3-3 (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein) proteins, which traps TFEB in the cytosol. Inactivation of MTORC1 leads to the transport of TFEB to the nucleus, thus stimulating the transcription of autophagy-associated genes [113,114]. Ser122 is another MTORC1-dependent phosphorylation site. The phosphorylation mimetic mutation Ser122Asp reduces nuclear TFEB when Ser211 is dephosphorylated but Ser122 dephosphorylation is not sufficient, in itself, to result in the nuclear localization of TFEB [115], indicating that Ser122 coordinates with Ser211 to control TFEB localization. Ser138 and Ser142 are localized in proximity to the nuclear export signal/NES on TFEB, and MTORC1-dependent phosphorylation at these two sites is critical for TFEB nuclear export [116]. Besides the direct regulation of TFEB, MTORC1 inhibits TFEB activity through activating KAT2B/GCN5, an acetyltransferase that acetylates TFEB and inhibits its DNA-binding activity [117]. Therefore, under nutrient stress, MTORC1 inhibition stimulates not only TFEB accumulation in the nucleus but its binding to target genes as well, thus promoting lysosomal biogenesis and autophagy flux.
TFE3 and MITF are additional TFs that drive the expression of genes involved in lysosome biogenesis and autophagy [118,119,120]. Similar to TFEB, TFE3 and MITF are recruited to lysosomes by activated RRAG GTPase, and MTORC1 inactivation during starvation is necessary for their release from YWHA/14-3-3 proteins and nuclear localization [111,118]. Ser321 on TFE3 is apparently an MTORC1-dependent phosphorylation site because both MTORC1 inactivation and Ser321Ala mutation abolish its interaction with YWHA/14-3-3 proteins and stimulate nuclear localization [118]. On MITF, Ser280, a residue that corresponds to TFEB Ser211 by homology analysis, is also nominated as a potential MTORC1 phosphorylation site [111]. Interestingly, a study found that TFEB and TFE3 positively regulate MTORC1 activity by promoting RRAGD expression and recruiting MTORC1 to the lysosome when nutrients are provided to the starved cells. Even though it remains as an open question as to how modulating RRAGD expression is sufficient for regulating MTORC1 activity, this mechanism may be important for cells to prepare for nutrient refeeding during starvation [121].
In addition to directly phosphorylating TFs and affecting their localization, MTORC1 also regulates the EIF2A (eukaryotic translation initiation factor 2A)-ATF4 pathway, which induces the expression of ATG genes [122,123]. During nutrient deprivation, MTORC1 inactivation induces PPP6C (protein phosphatase 6 catalytic subunit) phosphatase activity, which dephosphorylates and activates EIF2AK4/GCN2. EIF2AK4/GCN2 further phosphorylates and activates EIF2A, leading to the subsequent increase of ATF4 expression, ATG gene transcription induction and activated autophagy [122].
Third, MTORC1 is responsible for the posttranscriptional regulation of ATG genes. Recently, MTORC1 is reported to regulate autophagy via controlling mRNA N6-methyl-adenosine (m6A) methylation. In this study, the researchers found that MTORC1 activates the CCT (chaperonin containing TCP1) complex, which helps in the folding of proteins in the m6A methyltransferase complex, resulting in more m6A RNA methylation, the degradation of ATG transcripts and the suppression of autophagy [124].
Finally, in recent years epigenetics has been proposed to be an important regulatory aspect of autophagy [125]. Even though not much is known about the regulation of histone modifications by MTORC1 during nutrient stress, some studies provide initial clues. One example is that during starvation or rapamycin treatment, acetylation of histone H4 Lys16/H4K16ac and the corresponding acetyltransferase KAT8/hMOF are both downregulated, which is important for cell survival during starvation although the detailed mechanism is not known [80]. In addition, MTORC1 enhances the nuclear localization of FOXK1 and FOXK2, which recruit the SIN3A-HDAC complex to restrict the acetylation of histones and the expression of ATG genes [126]. These two examples suggest a close connection between MTORC1 and the epigenetic regulation of autophagy, but more studies are still needed to better understand this relationship.

2.2.2. Other Autophagy Regulation during Nutrient Stress

In addition to MTORC1, the main sensor of nutrients, several other stress-response kinases regulate autophagy during nutrient stress. For instance, the stress-activated signaling molecule MAPK8/JNK1 phosphorylates BCL2 during starvation, which prevents its interaction with BECN1, thus promoting autophagy [127]. MAPKAPK2 and MAPKAPK3, which belong to the stress-response kinase MAPK family, phosphorylates BECN1 Ser90 during starvation and this phosphorylation is important for BECN1 function [128]. Additionally, the IKK complex gets activated by starvation and induces the expression of several autophagy genes, therefore stimulating autophagy [129]. As mentioned above, multiple stress-response kinases contribute to the stimulation of autophagy during nutrient starvation, but whether, and how, these signaling pathways coordinate to regulate autophagy requires further attention.

3. Autophagy Regulation under Energy Stress

Recycling by autophagy is essential for yeast and mammals to survive starvation. The breakdown products and materials can be further used to provide building blocks for the synthesis of essential proteins and to produce ATP through catabolic pathways. Therefore, autophagy is essential for the maintenance of energy homeostasis and is finely regulated upon energy deprivation.
AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine protein kinase [130], sensing low cellular ATP levels and controlling turnover of cellular materials and metabolism, thus being essential for cellular adaptation to energy limitation [131]. AMPK is a heterotrimeric complex composed of a catalytic subunit (PRKAA/α) and two regulatory subunits (PRKAB/β and PRKAG/γ). The PRKAA-subunit contains the kinase domain and the critical residue Thr172 whose phosphorylation by upstream kinases activates AMPK activity [132]. The PRKAG-subunit possesses four cystathionine β-synthase/CBS motifs that can bind to all forms of adenosine-containing ligands, enabling it to sense the changes in the ATP:AMP/ADP ratio [133,134], and the activity of AMPK is precisely regulated by these ratios in the cell [130]. When cells are in the fed state, AMPK is mostly bound by ATP and its activity is inhibited. Under energy-starvation conditions, the cellular concentration of ATP decreases whereas levels of ADP and AMP increase. AMP or ADP binding to the PRKAG-subunit activates the kinase through three distinct mechanisms: (1) it promotes the STK11/LKB1 (serine/threonine kinase 11)-mediated phosphorylation of the PRKAA subunit at Thr172, which can increase AMPK activity up to 100-fold in vitro [135,136]; (2) it protects phosphorylated Thr172 from dephosphorylation by phosphatases [137]; and (3) it causes allosteric activation of the AMPK complex [138]. Once activated, AMPK serves as a central metabolic regulator to restore energy homeostasis by inhibiting anabolic pathways and promoting catabolic pathways, including autophagy. AMPK promotes autophagy at various steps by phosphorylating autophagy-related proteins or autophagy regulators.
Activated AMPK can induce the autophagic process by inhibiting the activity of MTOR in two ways (Figure 3): (1) AMPK directly phosphorylates the MTORC1 component RPTOR on Ser722 and Ser792. This phosphorylation induces YWHA/14-3-3 binding to RPTOR, thus hindering the binding of RPTOR to MTOR and MTOR substrates, leading to suppression of MTORC1 activity [139]. (2) AMPK phosphorylates the MTOR upstream regulator TSC2 on Thr1227 and Ser1345, which promotes the GTPase-activating function of the TSC1-TSC2 complex, leading to the transformation of RHEB into an inactive RHEB-GDP state, which consequently reduces MTOR activity [140,141]. As mentioned above, reduced MTOR activity relieves the inhibition on ULK1 to activate autophagy.
AMPK can also stimulate autophagy through phosphorylating autophagy related proteins including ULK1, BECN1, and PIK3C3/VPS34 (Figure 3). Under energy-starvation conditions, AMPK directly activates ULK1 through phosphorylation of Ser317, Ser467, Ser555, Ser574, Ser637, and Ser777 [98]. This activation is prevented by MTOR activity during normal physiological conditions as MTORC1 phosphorylates ULK1 at Ser757, which is located in the AMPK-ULK1 binding region (amino acids 711-828), thereby inhibiting the interaction between AMPK and ULK1 [98]. AMPK also regulates the PIK3C3/VPS34 lipid kinase complex upon glucose withdrawal: AMPK activates the pro-autophagy PIK3C3/VPS34 complex by phosphorylating Ser91 and Ser94 in BECN1, which increases autophagosome formation. In the meantime, AMPK inhibits the PIK3C3/VPS34 complexes not involved in autophagy by phosphorylating Thr163 and Ser165 in PIK3C3/VPS34 to suppresses overall PtdIns3P production. The presence of ATG14 dictates the differential regulation by inhibiting PIK3C3/VPS34 phosphorylation and increasing BECN1 phosphorylation by AMPK during glucose starvation [142]. Furthermore, AMPK can phosphorylate other core components of the autophagy pathway. For example, activated AMPK can phosphorylate ATG9A at Ser761, which recruits ATG9A to LC3-positive autophagosomes and enhances autophagosome production [143].
Apart from direct phosphorylation of the core components of the autophagy machinery, AMPK can also promote autophagy through activating autophagy regulators (Figure 3). For example, human transcription factor FOXO3 is phosphorylated by AMPK at Thr179, Ser399, Ser413, Ser555, Ser588, and Ser626, which promotes the nuclear translocation of FOXO3 and its activity, thus upregulating the transcription of downstream auto-phagy-related genes such as ATG4, ATG12, BECN1, LC3, and ULK1 [144,145]. The NAD-dependent deacetylase SIRT1 (sirtuin 1), an essential regulator of autophagy during energy deprivation, is also under the regulation of AMPK. Under glucose starvation conditions, cytoplasmic GAPDH is phosphorylated by activated AMPK and redistributes into the nucleus, where it interacts with SIRT1 and displaces SIRT1′s repressor CCAR2/DBC1 leading to the activation of SIRT1 [146]. SIRT1 can also be activated by the increased level of NAD+ during starvation. The targets of SIRT1 include, but are not limited to, autophagy pathway components ULK1, ATG5, and LC3 and the transcription factor FOXO1, which induces the expression of the GTPase RAB7 that mediates the fusion of autophagosomes with lysosomes [80,147,148]. During energy stress, a considerable amount of AMPK is translocated from the cytosol to mitochondrial-associated ER membrane/MAM, where it interacts with and phosphorylates the mitochondrial fusion protein MFN2. This AMPK-MFN2 axis is required for mitochondrial-associated ER membrane dynamics and auto-phagy induction [149].

4. Autophagy Regulation under Oxidative/Nitrosative Stress

Reactive oxygen and nitrogen species (hereafter ROS and RNS) are highly reactive molecules that can cause oxidative damages on macromolecules and biological membranes [150,151]. Cells have developed very sophisticated mechanisms to regulate the homeostasis of ROS and RNS, including endogenous antioxidants, such as glutathione and TXN (thioredoxin), and detoxifying enzymes, such as GPX (glutathione peroxidase), CAT (catalase), and SOD (superoxide dismutase), to efficiently resolve the excessive oxidative stress [151,152]. In coordination with the ubiquitin–proteasome system, autophagy plays essential roles in sequestering oxidized proteins in the lysosome/vacuole for degradation to maintain homeostasis [153,154,155]. However, autophagy is also involved in oxidative stress-induced cell death [156]. For example, the free iron released by ferritinophagy could promote lipid ROS accumulation, thus triggering ferroptosis and the increased autophagic flux in SOD1G93A transgenic lead to muscle atrophy [157,158,159]. To date, there is abundant evidence showing that the autophagy activity is tightly regulated by the oxidative stress [154,160,161,162].
As for the ROS, the direct reaction between oxygen and extra electron gives rise to the superoxide (O2●−), which is highly reactive and is rapidly converted into hydrogen peroxide by the endogenous SOD [163]. H2O2 is relatively stable and is considered as an important signaling molecule for the ROS responsive pathways [164,165]. In the presence of iron, H2O2 can generate the unstable hydroxyl radical (HO) via a process called the Fenton reaction. Hydroxyl radicals can further react with polyunsaturated fatty acid to form various lipid peroxides [166,167]. Autophagy is activated by each of these different ROS species as well as numerous RNS species [156,168,169,170,171,172,173,174,175,176].
In this section, we cover the current understanding of the regulatory mechanisms of autophagy in yeast and mammalian cells. The relationship between mitophagy and oxidative stress has been reviewed by De Gaetano et al., in the same special issue [177].

4.1. Mechanisms of Autophagy Regulation by Oxidative Stress in Yeast

The regulatory role of oxidative stress on autophagy is evolutionarily conserved in the budding yeast Saccharomyces cerevisiae. For example, yeast mitophagy induced by nitrogen starvation and ethanol challenge both can be prevented by adding the antioxidant N-acetylcysteine/NAC [178,179].
Yap1 signaling is the most well-characterized oxidative stress responsive pathway in yeast [180,181]. Yap encompasses a transcription factor family of eight basic leucine zipper (bZIP) domain proteins [182,183]. Among them, Yap1 can directly translocate into the nucleus to activate the expression of various antioxidant genes such as TRX2 by the stimulation of oxidative stress [184,185]. Under basal conditions, Yap1 is enriched in the cytosol as the nuclear exporter Crm1 efficiently pumps Yap1 out of the nucleus [185]. Upon H2O2 activation, however, Yap1 is oxidized, and several disulfide bonds are formed on its C-terminal cysteine-rich domain/c-CRD and amino-terminal cysteine-rich domain/n-CRD so that the Crm1-cognate nuclear export signal is masked. As a result, Yap1 is trapped in the nucleus where it activates the expression of stress-responsive genes [186,187]. The oxidation of Yap1 (especially the covalent bonds between Cys303 and Cys598) requires the participation of the thiol peroxidase Hyr1/Gpx3/Orp1, which acts as a direct receptor for H2O2 [188]. Yap1 recognizes a consensus DNA element in the promoter region called the Yap response element (YRE, which includes TGACTAA, TTAGTCA, TTACTAA, and T[T/G]ACAAA) [180]. Among all currently known ATG genes, only ATG15 contains a potential binding site for Yap1. ATG15 encodes a vacuolar phospholipase that can break down the inner autophagosome membrane in the vacuole lumen, and its direct activation by Yap1 has been experimentally verified (Figure 4A) [189,190].
Atg4 is a cysteine protease, and the mammalian homolog has been reported to be directly regulated by H2O2 (see below for further details) [191]. However, the Cys81 residue on human ATG4A and ATG4B that is proposed to play important roles in this process is not conserved in yeast. Yeast Atg4 is also redox regulated through a different mechanism: site-directed mutagenesis reveals that a single disulfide bond formed by Cys338 and Cys394 has a very low redox potential and is required for Atg4 redox regulation in yeast; the formation of this disulfide bond decreases the Atg4 protease activity and can be rapidly reduced by thioredoxin [192].

4.2. Mechanisms of Autophagy Regulation by Oxidative Stress in Mammalian Cells

In mammalian cells, ROS accumulation can be triggered by several different stimuli, such as hypoxia, nutrient stress or cytokines including TNF/TNFα [193,194,195,196,197]. Therefore, many upstream signaling pathways have been proposed to affect the activity of auto-phagy, including signaling by NFKB/NF-κB, AMPK, HIF1A/HIF-1, ATM, AKT-MTOR, and MAPK [194,195,197,198,199,200,201,202,203,204]. The regulation of autophagy by these upstream signaling pathways has been well summarized elsewhere [153,154,160,205].
The NFE2L2/Nrf2 (NFE2 like2 bZIP transcription factor 2)-KEAP1 (kelch ECH associated protein 1)-antioxidant signaling pathway can be directly activated by oxidative stress and can stimulate the expression of various stress-responsive genes including several detoxifying enzymes and autophagy proteins [206,207,208]. As a functional ortholog for Yap1, NFE2L2 is also a bZIP transcription factor; heterodimers of NFE2L2 and MAF proteins recognize a specific antioxidant response element/ARE in the promoter region of target genes [209,210]. Under basal conditions, the NFE2L2 is localized in the cytoplasm and maintained at a very low level. This dynamic regulation is achieved by its interacting partner KEAP1 which is a CUL3 (cullin 3) E3 ubiquitin ligase adaptor [211,212,213]. KEAP1 interacts with NFE2L2 via the carboxy-terminal Kelch domain with a 2:1 stoichiometry. At the same time, KEAP1 interacts with the CUL3 ligase via the amino-terminal bric-a-brac, tramtrack, and broad complex/BTB domain, thus promoting the efficient degradation of NFE2L2 [211,214,215,216]. KEAP1 acts as the redox sensor and the interaction between KEAP1 and NFE2L2 is directly regulated by environmental cues via a mechanism called the “hinge and latch model”: in response to H2O2, KEAP1 Cys226, Cys613, and Cys622/624 residues form disulfide bonds that impair the interaction between KEAP1-CUL3 and NFE2L2, thus stabilizing NFE2L2 and releasing it into the nucleus where it is active [212,213,217,218,219].
The SQSTM1/p62 (sequestosome 1) protein contains an LC3-interacting region (LIR) domain thus allows it to act as an autophagy receptor to facilitate delivery of cargos into the phagophore for subsequent degradation [220,221]. The antioxidant response element has been identified in the promoter of SQSTM1 that makes it a target for NFE2L2 activation [222]. Interestingly, KEAP1 is among the autophagic substrates of SQSTM1. Therefore, SQSTM1-mediated autophagy can degrade KEAP1 to further activate NFE2L2 signaling in a positive feedback loop [223,224,225,226]. Recently, more NFE2L2-targeted autophagy genes have been reported, including ULK1, CALCOCO2, ATG4D, ATG7, GABARAPL1, ATG2B, ATG5, and LAMP2B, suggesting that, in addition to TFEB and FOXO, NFE2L2 is an important autophagy regulator, perhaps in a more oxidative-stress relevant context (Figure 4B) [227,228].
In addition, ROS can regulate autophagy by directly oxidizing the cysteine residues on the core autophagy components. For example, the cysteine protease ATG4A and ATG4B can be inactivated by H2O2. The possible mechanism is that Cys81 is sensitive to oxidation, triggering a conformation change on Cys77 which is the catalytic residue, thus inhibiting the cysteine protease activity of ATG4 [191]. As a result, the transient blockade of ATG4 activity stabilizes the lipidated forms of its substrates LC3 and GABARAPL2/GATE-16 so that autophagosome biogenesis is promoted [191]. Of note, this reversible inhibition of ATG4 is spatially and temporally regulated. As the autophagosome is trafficked towards lysosomes where the local H2O2 is lower, ATG4 is reactivated to deconjugate and recycle the LC3 and GABARAPL2. Similarly, in the context of mito-phagy, the cysteine residues of the ubiquitin E3 ligase PRKN/parkin can be oxidized by sulfhydration that is required for full PRKN ligase activity and normal mitophagy flux [229,230].

4.3. Mechanisms of Autophagy Regulation by Nitrosative Stress

In both yeast and mammals, nitric oxide (NO) mediates critical physiological functions as a signaling molecule at low concentrations, but causes nitrosative stress at high concentrations [231,232,233]. Imbalance of reactive nitrogen species (RNS) results in accumulation of protein tyrosine nitration, protein S-nitrosylation on cysteine residues, and damage to lipids and DNA [234,235]. In mammalian systems, nitrosative stress is correlated with many pathological conditions, such as cancer, neurodegenerative diseases, and ischemia, and upregulated autophagy activity is observed in several nitrosative stress models [236,237,238]. For example, in a microsphere embolism rat model, the increased autophagy signaling (protein level of BECN1, LC3, LAMP2, and CTSB [cathepsin B]) is accompanied by nitrosative stress, which can be partially resolved by adding the peroxynitrite (ONOO) scavenger melatonin [239]. Furthermore, RNS can attenuate MTORC1 activity to promote autophagy via the ATM-AMPK-TSC2 and AKT signaling axis [240,241]. A recent study challenged MCF7 cells with the NO donor compound DETA-NONOate and observed an increased NAD+:NADH ratio. The pharmacological and genetic inhibition of the NAD+-dependent deacetylase SIRT1 reduces autophagy activity, and the acetylation of TP53, and promotes cell survival, suggesting the complex interplay among SIRT1, TP53, and autophagy upon nitrosative stress (Figure 4C) [242]. However, whether autophagy is required for RNS homeostasis will requires further loss-of-function studies of autophagy genes and their products.

5. Autophagy Regulation under ER Stress

5.1. Endoplasmic Reticulum Stress and Autophagy

The endoplasmic reticulum (ER) is a central membrane-bound organelle, and its membrane structure was first documented by Porter et al. in 1945 using electron microscopy [243]. The ER is an important organelle in eukaryotic cells with various functions, such as protein synthesis, modification and processing of proteins, secretion of correctly folded proteins, calcium homeostasis, and lipid and carbohydrate metabolism [244,245]. Therefore, the ER is essential for cell homeostasis.
Normally, the ER utilizes chaperones to properly fold newly synthesized proteins and identify misfolded proteins for destruction. However, the ER homeostasis is disrupted under numerous pathological conditions including nutrient deprivation, perturbation of cellular ATP level, calcium metabolic imbalance, redox imbalance, viral infection, and the presence of environmental toxins. In addition, the protein-folding capacity of the ER can also be compromised and eventually cause the accumulation of unfolded or misfolded proteins in the ER lumen, also known as ER stress [246].
The ER stress triggers an adaptive response referred to as the unfolded protein response (UPR). After sensing the ER stress, the UPR transduces the signal to the regulation of downstream transcription factors and then induces ER chaperone genes to upregulate the folding capacity. Additionally, the cells can also begin a process termed ER-associated degradation (ERAD) to mediate the transport of unfolded or misfolded proteins into the cytosol for degradation. ERAD mainly consists of two mechanisms: ubiquitin–proteasome-dependent ERAD/ERAD(I) and autophagy–lysosome dependent ERAD/ERAD(II) [247,248,249]. ER-to-lysosome-associated degradation/ERLAD is the name currently used for the autophagy-dependent mechanism that is employed to handle proteins that cannot be degraded by ERAD.
As mentioned in the previous section in this review, autophagy can be induced by different types of cellular stress, which includes the ER stress discussed here. The relationship between ER stress and autophagy was first described in 2006 in yeast [250,251]. Here, we name the autophagy activated by ER stress as “ER stress-mediated autophagy” because this is the term used in most studies.

5.2. The Mechanisms of ER Stress-Mediated Autophagy in Yeast

In yeast, the ER stress can be sensed by an ER-resident type 1 transmembrane protein called Ire1, which plays a critical role in the UPR induced by ER stress [252,253]. It is noteworthy that Ire1 was initially identified as an mRNA splicing factor in yeast [254,255]. In addition, Ire1 is also capable of sensing unfolded or misfolded proteins in the ER lumen because Ire1 has both an endoribonuclease domain and an ER lumenal stress-sensing domain.
Ire1 is localized in the ER membrane with its C terminus facing into the cytosol and the N terminus residing in the ER lumen. Under normal conditions, the N-terminal region of Ire1 is bound to Kar2 unless Ire1 senses the accumulation of unfolded proteins in the ER lumen. Ire1 is activated by autophosphorylation after dissociation from Kar2, leading to the expression of activated Hac1, a transcription factor.
Upon the UPR induced by ER stress, a non-classical intronic sequence near the 3’ end of the open reading frame of HAC1 mRNA is excised by activated Ire1 and then the two ends of the mRNA are ligated by the tRNA ligase Trl1 [256]. The spliced HAC1 encodes an activated form of the Hac1 protein containing 238 amino acids, which contains 18 amino acids more than the Hac1 protein encoded by un-spliced HAC1 mRNA; these 18 amino acids play a key role for TF activation [257]. The difference in the properties of the two types of Hac1 protein is mainly caused by the C terminus. The N terminus of both types of Hac1 has a DNA-binding function while the C terminus of activated Hac1 has an active transcriptional activation domain due to the cleavage and splicing reaction [258]. Eventually, activated Hac1 is exported to the nucleus and binds to the unfolded protein response elements/UPREs to promote the transcription of UPR-related genes [259]. Furthermore, studies have reported that these unfolded protein response elements are commonly found in the promoters of some UPR-related genes including FPR2/FKB2, KAR2, and PDI1 [260].
Studies carried out in 2006 show that ER stress can induce autophagy through this Ire1-Hac1 pathway in yeast [250,251]. Yorimitsu et al. used two types of drugs: dithiothreitol/DTT (an inhibitor of disulfide bond formation) and tunicamycin (an inhibitor of glycosylation) to induce ER stress. Then GFP-Atg8 processing and precursor Ape1 maturation assays were applied to monitor autophagic induction after the drug treatment. Both assays showed increased autophagic flux, indicating an induction of autophagy caused by ER stress. Additionally, the necessity of the Ire1-Hac1 signaling pathway for this ER stress-mediated autophagy was also explored in this study. The authors found that deletion of either IRE1 or HAC1 does not affect the capability for inducing autophagy caused by nutrient depletion; however, either knockout did block ER stress-mediated autophagy, suggesting that Ire1 and Hac1 are involved in the induction of this pathway probably through the UPR.

5.3. The Mechanisms of ER Stress-Mediated Autophagy in Mammalian Cells

The UPR is a highly conserved mechanism and mammalian cells also utilize it to alleviate ER stress by enhancing the protein-folding capacity of, and reducing the protein synthetic load on this organelle to restore ER homeostasis [259]. Unlike the UPR in yeast, that consists of the Ire1 signaling pathway, the UPR in mammalian cells is characterized by three major branches involving three ER membrane resident proteins: the serine/threonine-protein kinase/endoribonuclease ERN1/IRE1α (endoplasmic reticulum to nucleus signaling 1), EIF2AK3/PERK (eukaryotic translation initiation factor 2 alpha kinase 3), and the cyclic AMP-dependent transcription factor ATF6 (activating transcription factor 6).
Under normal physiological conditions, all three ER stress sensors are inactive due to the binding of an ER-resident chaperone protein, HSPA5/BIP/GRP78. Due to the high affinity of unfolded or misfolded proteins for HSPA5, ERN1, EIF2AK3, and ATF6 become active when there is an accumulation of unfolded or misfolded proteins in the ER lumen [261]. Moreover, the increased activity of these three ER sensors is also partially contributed to by unfolded or misfolded proteins acting as active ligands for their activation [262]. The activation of these three UPR signaling pathways alleviates ER stress by partially overlapping but distinct mechanisms, including autophagy.

5.3.1. ERN1

ERN1 is a bifunctional protein in mammalian cells consisting of three domains: an N-terminal lumenal domain, a cytosolic endoribonuclease domain, and a cytosolic serine/threonine kinase domain [263]. Similar to the Ire1 in yeast, active ERN1 can excise a 26-nucletide intron from XBP1 mRNA. The spliced XBP1 mRNA allows the expression of an active and stable form of XBP1 (X-box binding protein 1). The transcription factor XBP1 is then translocated to the nucleus and upregulates the expression of target genes in response to ER stress [264]. Among the target genes, BECN1 plays a central role in autophagy, suggesting that the splicing of XBP1 mRNA mediated by ERN1 under ER stress is important for autophagy induction [265]. Consistently, studies report that the un-spliced XBP1 mRNA can interact with FOXO1 (forkhead box O1), resulting in a decreased level of this TF, finally leading to the downregulation of autophagy [266,267,268].
Additionally, ERN1 can interact with TRAF2 (TNF receptor associated factor 2) and form a complex, which can phosphorylate MAP3K5/ASK1 (mitogen-activated protein kinase kinase kinase 5). Next, the phosphorylated MAP3K5 actives MAPK8/JNK1 (mitogen-activated protein kinase 8) by phosphorylation. Subsequently, the phosphorylated MAPK8-mediated phosphorylation of BCL2 can increase the level of free BECN1 by disrupting the BECN1-BCL2 complex, or by elevating BECN1 transcription, which leads to autophagosome formation [127,269]. Moreover, it is reported that the activation of AMPK mediated by ERN1 is involved in autophagy initiation [270].

5.3.2. EIF2AK3

Under ER stress conditions, the activation of the EIF2AK3 UPR signaling pathway upregulates many autophagy-related genes. The active EIF2AK3 can mediate the phosphorylation of EIF2A, which can elevate both ATG12 mRNA and protein levels [271]. In addition, EIF2AK3-mediated EIF2A phosphorylation also enables the selective translation of ATF4 mRNA, and the transcription factor ATF4 is then translocated to the nucleus where it upregulates the expression of multiple proteins, such as several autophagy-related proteins (ATG3, ATG12, ATG16L1, BECN1, and LC3) and DDIT3/CHOP (DNA damage inducible transcript 3) [272]. The expression of DDIT3 can also transcriptionally increase the expression of some proteins involved in autophagy (ATG5, ATG10, and GABARAP). In addition, DDIT3 can downregulate the expression of BCL2, a protein that binds to BECN1 and inhibits autophagosome formation [273,274]. Interestingly, the complex formed by ATF4 and DDIT3 can also induce the expression of some proteins involved in autophagy, including ATG7, NBR1 (NBR1 autophagy cargo receptor), and SQSTM1 [123]. Furthermore, the active EIF2AK3 pathway can initiate autophagy via the activation of AMPK and the inhibition of MTORC1 [275]. Consistent with this finding, the activation of ATF4-DDIT3 mediated by EIF2AK3 inhibits MTORC1 activity resulting in the induction of autophagy [276].

5.3.3. ATF6

Under ER stress conditions, ATF6 is translocated to the Golgi apparatus where it is cleaved by MBTPS1/S1P and MBTPS2/S2P. The N-terminal domain of ATF6 after the cleavage is translocated to the nucleus to induce the expression of UPR genes, including DDIT3 and XBP1 [277,278,279]. Therefore, ATF6 can indirectly regulate autophagy through the DDIT3 and XBP1 signaling pathway as mentioned above. In addition, ATF6 might regulate autophagy in the initiation step by the inhibition of AKT activity [280]. Additionally, ATF6 can interact with the transcription factor CEBPB (CCAAT enhancer binding protein beta) and then stimulate the expression of DAPK1 (death associated protein kinase 1) [281,282], which can phosphorylate BECN1 so that it will be released from the auto-phagy inhibitory BECN1-BCL2 complex, promoting the induction of autophagy.

5.3.4. Calcium

The ER is a multifunctional organelle which plays a pivotal role in maintaining intracellular calcium homeostasis. Under ER stress conditions, the calcium homeostasis is disrupted and the release of calcium from the ER to the cytosol is also elevated, which can induce autophagy. When calcium is released from the ER through ITPR (inositol 1,4,5-trisphosphate receptor), the CAMKK-AMPK-dependent signaling pathway is activated and the inhibitory effect of MTOR on the ULK1 complex is relieved [283,284]. Moreover, calcium release can activate DAPK1 [285], which is involved in the induction step of autophagy as noted above.

6. Conclusions

In this review, we summarized autophagy regulation under different types of stress, including that involving nutrients, energy, oxidation, and the ER (Figure 5). Besides the ones mentioned in this review, other types of stress, such as DNA damage and pathogen infection, are also able to induce autophagy [286,287]. The fact that autophagy is induced by multiple stresses highlights the importance of autophagy in allowing cells to maintain homeostasis in response to changes in the environment.
Of note, in this review, we mainly focus on how stress-response molecules and/or pathways regulate autophagy. There are a wide range of regulatory mechanisms affecting autophagy-associated genes under stress conditions, especially during nutrient deprivation, but the connections with stress-sensing pathways have not been established [288]. Additionally, apart from stress, the activity of stress-sensing molecules can also be regulated by other factors, and all these contribute to autophagy regulation under stress conditions. We did not discuss these factors in detail because they are beyond the scope of this review, but their roles in autophagy regulation cannot be ignored.
Even though we introduced different types of stress separately, it does not mean that the stress-responding molecules or pathways function alone. In fact, stress-responding pathways have very close connections, regulating each other or sharing the same downstream effector. Using the nutrient sensor MTORC1 and energy sensor AMPK as an example, it is well known that AMPK inhibits MTORC1 and both kinases target ULK1 to regulate its activity [53,98]. The regulatory network among AMPK, ULK1, and MTORC1 are important for the oscillation of autophagy [289]. Recently, AMPK was shown to be inhibited by MTORC1 [290], further indicating the complex interaction between these two important stress responders. Another example is seen with EIF4A, which is activated by multiple stresses and induces the expression level of ATF4, a transcription factor that promotes the transcription of multiple ATG genes [272]. Similarly, NFE2L2 is also in the center of the stress response, as the expression of this transcription factor is activated not only by oxidative stress, but also by other conditions such as ER stress [291]. The interaction between different stress-responding pathways and the existence of a common response pathway makes autophagy induction by stress a rapid and well-controlled process.
Thanks to advanced studies in recent decades, we can now draw a clearer picture of the autophagy regulation network under stress conditions. However, more studies focusing on this field are still needed for a better understanding on how autophagy is controlled, because too much or too little autophagy can harm cells. More importantly, the insights on autophagy regulation under stress may shed light on understanding the relation between autophagy and disease because cells in a diseased state usually undergo stress. For example, cancer cells in the interior of a tumor usually experience nutrient and oxidative stress because of the lack of proximal blood vessels. ER stress and oxidative stress are also proposed to contribute to neurodegenerative diseases [292,293]. Autophagy has a close connection with these diseases [18], therefore, a deeper understanding of autophagy regulation under stress conditions may help us find more potential autophagy-targeting therapeutic approaches.

Author Contributions

Preparation, writing and editing of the manuscript, Y.L., Y.H., X.W., Z.Y., Z.Z., D.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institutes of Health grant number GM131919.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMPKAMP-activated protein kinase
ATGautophagy related
ERendoplasmic reticulum
ERADER-associated degradation
GAPGTPase activating protein
H2O2hydrogen peroxide
m6AN6-methyl-adenosine
MTORC1mechanistic target of rapamycin kinase complex 1
NOnitric oxide
PASphagophore assembly site
PEphosphatidylethanolamine
PtdIns3Kclass III phosphatidylinositol kinase
RNSreactive nitrogen species
ROSreactive oxygen species
TFstranscription factors
Ublubiquitin-like
UPRunfolded protein response
V-ATPasevacuolar-type H+-translocating ATPase

References

  1. Yorimitsu, T.; Klionsky, D.J. Autophagy: Molecular machinery for self-eating. Cell Death Differ. 2005, 12 (Suppl. 2), 1542–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Wen, X.; Klionsky, D.J. An overview of macroautophagy in yeast. J. Mol. Biol. 2016, 428, 1681–1699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yang, Z.; Klionsky, D.J. An overview of the molecular mechanism of autophagy. Curr. Top. Microbiol. Immunol. 2009, 335, 1–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [Green Version]
  5. Nakatogawa, H. Mechanisms governing autophagosome biogenesis. Nat. Rev. Mol. Cell Biol. 2020, 21, 439–458. [Google Scholar] [CrossRef]
  6. Yim, W.W.; Mizushima, N. Lysosome biology in autophagy. Cell Discov. 2020, 6, 6. [Google Scholar] [CrossRef] [Green Version]
  7. Ariosa, A.R.; Lahiri, V.; Lei, Y.; Yang, Y.; Yin, Z.; Zhang, Z.; Klionsky, D.J. A perspective on the role of autophagy in cancer. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166262. [Google Scholar] [CrossRef]
  8. Ohsumi, Y. Historical landmarks of autophagy research. Cell Res. 2014, 24, 9–23. [Google Scholar] [CrossRef] [Green Version]
  9. Gatica, D.; Chiong, M.; Lavandero, S.; Klionsky, D.J. The role of autophagy in cardiovascular pathology. Cardiovasc. Res. 2022, in press. [Google Scholar] [CrossRef]
  10. Suzuki, K.; Kirisako, T.; Kamada, Y.; Mizushima, N.; Noda, T.; Ohsumi, Y. The pre-autophagosomal structure organized by concerted functions of APG genes is essential for autophagosome formation. EMBO J. 2001, 20, 5971–5981. [Google Scholar] [CrossRef]
  11. Mizushima, N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr. Opin. Cell Biol. 2010, 22, 132–139. [Google Scholar] [CrossRef] [PubMed]
  12. Burman, C.; Ktistakis, N.T. Regulation of autophagy by phosphatidylinositol 3-phosphate. FEBS Lett. 2010, 584, 1302–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Geng, J.; Klionsky, D.J. The Atg8 and Atg12 ubiquitin-like conjugation systems in macroautophagy. ‘Protein modifications: Beyond the usual suspects’ review series. EMBO Rep. 2008, 9, 859–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Shpilka, T.; Mizushima, N.; Elazar, Z. Ubiquitin-like proteins and autophagy at a glance. J. Cell Sci. 2012, 125, 2343–2348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Yin, Z.; Popelka, H.; Lei, Y.; Yang, Y.; Klionsky, D.J. The Roles of Ubiquitin in Mediating Autophagy. Cells 2020, 9, 2025. [Google Scholar] [CrossRef]
  16. Ichimura, Y.; Kirisako, T.; Takao, T.; Satomi, Y.; Shimonishi, Y.; Ishihara, N.; Mizushima, N.; Tanida, I.; Kominami, E.; Ohsumi, M.; et al. A ubiquitin-like system mediates protein lipidation. Nature 2000, 408, 488–492. [Google Scholar] [CrossRef]
  17. Reggiori, F.; Tucker, K.A.; Stromhaug, P.E.; Klionsky, D.J. The Atg1-Atg13 complex regulates Atg9 and Atg23 retrieval transport from the pre-autophagosomal structure. Dev. Cell 2004, 6, 79–90. [Google Scholar] [CrossRef] [Green Version]
  18. Lei, Y.; Klionsky, D.J. The Emerging Roles of Autophagy in Human Diseases. Biomedicines 2021, 9, 1651. [Google Scholar] [CrossRef]
  19. Yu, X.; Long, Y.C.; Shen, H.M. Differential regulatory functions of three classes of phosphatidylinositol and phosphoinositide 3-kinases in autophagy. Autophagy 2015, 11, 1711–1728. [Google Scholar] [CrossRef] [Green Version]
  20. Nascimbeni, A.C.; Codogno, P.; Morel, E. Phosphatidylinositol-3-phosphate in the regulation of autophagy membrane dynamics. FEBS J. 2017, 284, 1267–1278. [Google Scholar] [CrossRef]
  21. Sun, Q.; Fan, W.; Chen, K.; Ding, X.; Chen, S.; Zhong, Q. Identification of Barkor as a mammalian autophagy-specific factor for Beclin 1 and class III phosphatidylinositol 3-kinase. Proc. Natl. Acad. Sci. USA 2008, 105, 19211–19216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Lu, J.; He, L.; Behrends, C.; Araki, M.; Araki, K.; Jun Wang, Q.; Catanzaro, J.M.; Friedman, S.L.; Zong, W.X.; Fiel, M.I.; et al. NRBF2 regulates autophagy and prevents liver injury by modulating Atg14L-linked phosphatidylinositol-3 kinase III activity. Nat. Commun. 2014, 5, 3920. [Google Scholar] [CrossRef] [PubMed]
  23. Fimia, G.M.; Stoykova, A.; Romagnoli, A.; Giunta, L.; Di Bartolomeo, S.; Nardacci, R.; Corazzari, M.; Fuoco, C.; Ucar, A.; Schwartz, P.; et al. Ambra1 regulates autophagy and development of the nervous system. Nature 2007, 447, 1121–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Polson, H.E.; de Lartigue, J.; Rigden, D.J.; Reedijk, M.; Urbe, S.; Clague, M.J.; Tooze, S.A. Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation. Autophagy 2010, 6, 506–522. [Google Scholar] [CrossRef] [Green Version]
  25. Klionsky, D.J.; Petroni, G.; Amaravadi, R.K.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cadwell, K.; Cecconi, F.; Choi, A.M.K.; et al. Autophagy in major human diseases. EMBO J. 2021, 40, e108863. [Google Scholar] [CrossRef]
  26. Gatica, D.; Lahiri, V.; Klionsky, D.J. Cargo recognition and degradation by selective autophagy. Nat. Cell Biol. 2018, 20, 233–242. [Google Scholar] [CrossRef]
  27. Urban, J.; Soulard, A.; Huber, A.; Lippman, S.; Mukhopadhyay, D.; Deloche, O.; Wanke, V.; Anrather, D.; Ammerer, G.; Riezman, H.; et al. Sch9 is a major target of TORC1 in Saccharomyces cerevisiae. Mol. Cell 2007, 26, 663–674. [Google Scholar] [CrossRef]
  28. Tanigawa, M.; Maeda, T. An In Vitro TORC1 Kinase Assay That Recapitulates the Gtr-Independent Glutamine-Responsive TORC1 Activation Mechanism on Yeast Vacuoles. Mol. Cell. Biol. 2017, 37, e00075-17. [Google Scholar] [CrossRef] [Green Version]
  29. Ikenoue, T.; Hong, S.; Inoki, K. Monitoring mammalian target of rapamycin (mTOR) activity. Methods Enzymol. 2009, 452, 165–180. [Google Scholar] [CrossRef]
  30. Zhang, C.S.; Hawley, S.A.; Zong, Y.; Li, M.; Wang, Z.; Gray, A.; Ma, T.; Cui, J.; Feng, J.W.; Zhu, M.; et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature 2017, 548, 112–116. [Google Scholar] [CrossRef]
  31. Berg, J.; Hung, Y.P.; Yellen, G. A genetically encoded fluorescent reporter of ATP:ADP ratio. Nat. Methods 2009, 6, 161–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tantama, M.; Martinez-Francois, J.R.; Mongeon, R.; Yellen, G. Imaging energy status in live cells with a fluorescent biosensor of the intracellular ATP-to-ADP ratio. Nat. Commun. 2013, 4, 2550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Zegzouti, H.; Zdanovskaia, M.; Hsiao, K.; Goueli, S.A. ADP-Glo: A Bioluminescent and homogeneous ADP monitoring assay for kinases. Assay Drug Dev. Technol. 2009, 7, 560–572. [Google Scholar] [CrossRef] [PubMed]
  34. Woods, A.; Munday, M.R.; Scott, J.; Yang, X.; Carlson, M.; Carling, D. Yeast SNF1 is functionally related to mammalian AMP-activated protein kinase and regulates acetyl-CoA carboxylase in vivo. J. Biol. Chem. 1994, 269, 19509–19515. [Google Scholar] [CrossRef]
  35. Lim, C.T.; Lolli, F.; Thomas, J.D.; Kola, B.; Korbonits, M. Measurement of AMP-activated protein kinase activity and expression in response to ghrelin. Methods Enzymol. 2012, 514, 271–287. [Google Scholar] [CrossRef]
  36. Hempel, S.L.; Buettner, G.R.; O’Malley, Y.Q.; Wessels, D.A.; Flaherty, D.M. Dihydrofluorescein diacetate is superior for detecting intracellular oxidants: Comparison with 2′,7′-dichlorodihydrofluorescein diacetate, 5(and 6)-carboxy-2′,7′-dichlorodihydrofluorescein diacetate, and dihydrorhodamine 123. Free Radic. Biol. Med. 1999, 27, 146–159. [Google Scholar] [CrossRef]
  37. Miller, E.W.; Tulyathan, O.; Isacoff, E.Y.; Chang, C.J. Molecular imaging of hydrogen peroxide produced for cell signaling. Nat. Chem. Biol. 2007, 3, 263–267. [Google Scholar] [CrossRef]
  38. Uggeri, J.; Gatti, R.; Belletti, S.; Scandroglio, R.; Corradini, R.; Rotoli, B.M.; Orlandini, G. Calcein-AM is a detector of intracellular oxidative activity. Histochem. Cell Biol. 2004, 122, 499–505. [Google Scholar] [CrossRef]
  39. Tian, R.; Abarientos, A.; Hong, J.; Hashemi, S.H.; Yan, R.; Drager, N.; Leng, K.; Nalls, M.A.; Singleton, A.B.; Xu, K.; et al. Genome-wide CRISPRi/a screens in human neurons link lysosomal failure to ferroptosis. Nat. Neurosci. 2021, 24, 1020–1034. [Google Scholar] [CrossRef]
  40. Camera, E.; Picardo, M. Analytical methods to investigate glutathione and related compounds in biological and pathological processes. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2002, 781, 181–206. [Google Scholar] [CrossRef]
  41. Owen, J.B.; Butterfield, D.A. Measurement of oxidized/reduced glutathione ratio. Methods Mol. Biol. 2010, 648, 269–277. [Google Scholar] [CrossRef] [PubMed]
  42. Dooley, C.T.; Dore, T.M.; Hanson, G.T.; Jackson, W.C.; Remington, S.J.; Tsien, R.Y. Imaging dynamic redox changes in mammalian cells with green fluorescent protein indicators. J. Biol. Chem. 2004, 279, 22284–22293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Meyer, A.J.; Dick, T.P. Fluorescent protein-based redox probes. Antioxid. Redox Signal. 2010, 13, 621–650. [Google Scholar] [CrossRef] [PubMed]
  44. Pap, E.H.; Drummen, G.P.; Winter, V.J.; Kooij, T.W.; Rijken, P.; Wirtz, K.W.; Op den Kamp, J.A.; Hage, W.J.; Post, J.A. Ratio-fluorescence microscopy of lipid oxidation in living cells using C11-BODIPY(581/591). FEBS Lett. 1999, 453, 278–282. [Google Scholar] [CrossRef] [Green Version]
  45. Dahle, L.K.; Hill, E.G.; Holman, R.T. The thiobarbituric acid reaction and the autoxidations of polyunsaturated fatty acid methyl esters. Arch. Biochem. Biophys. 1962, 98, 253–261. [Google Scholar] [CrossRef]
  46. Tang, Q.; Su, Y.W.; Xian, C.J. Determining Oxidative Damage by Lipid Peroxidation Assay in Rat Serum. Bio Protoc. 2019, 9, e3263. [Google Scholar] [CrossRef]
  47. Lajoie, P.; Fazio, E.N.; Snapp, E.L. Approaches to imaging unfolded secretory protein stress in living cells. Endoplasmic Reticulum Stress Dis. 2014, 1, 27–39. [Google Scholar] [CrossRef]
  48. Beriault, D.R.; Werstuck, G.H. Detection and quantification of endoplasmic reticulum stress in living cells using the fluorescent compound, Thioflavin T. Biochim. Biophys. Acta 2013, 1833, 2293–2301. [Google Scholar] [CrossRef] [Green Version]
  49. Iwawaki, T.; Akai, R.; Kohno, K.; Miura, M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat. Med. 2004, 10, 98–102. [Google Scholar] [CrossRef]
  50. Samali, A.; Fitzgerald, U.; Deegan, S.; Gupta, S. Methods for monitoring endoplasmic reticulum stress and the unfolded protein response. Int. J. Cell Biol. 2010, 2010, 830307. [Google Scholar] [CrossRef]
  51. Nadanaka, S.; Yoshida, H.; Kano, F.; Murata, M.; Mori, K. Activation of mammalian unfolded protein response is compatible with the quality control system operating in the endoplasmic reticulum. Mol. Biol. Cell 2004, 15, 2537–2548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Gonzalez, A.; Hall, M.N. Nutrient sensing and TOR signaling in yeast and mammals. EMBO J. 2017, 36, 397–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Gonzalez, A.; Hall, M.N.; Lin, S.C.; Hardie, D.G. AMPK and TOR: The Yin and Yang of Cellular Nutrient Sensing and Growth Control. Cell Metab. 2020, 31, 472–492. [Google Scholar] [CrossRef]
  54. Vlahakis, A.; Graef, M.; Nunnari, J.; Powers, T. TOR complex 2-Ypk1 signaling is an essential positive regulator of the general amino acid control response and autophagy. Proc. Natl. Acad. Sci. USA 2014, 111, 10586–10591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Stracka, D.; Jozefczuk, S.; Rudroff, F.; Sauer, U.; Hall, M.N. Nitrogen source activates TOR (target of rapamycin) complex 1 via glutamine and independently of Gtr/Rag proteins. J. Biol. Chem. 2014, 289, 25010–25020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Binda, M.; Peli-Gulli, M.P.; Bonfils, G.; Panchaud, N.; Urban, J.; Sturgill, T.W.; Loewith, R.; De Virgilio, C. The Vam6 GEF controls TORC1 by activating the EGO complex. Mol. Cell 2009, 35, 563–573. [Google Scholar] [CrossRef] [Green Version]
  57. Panchaud, N.; Peli-Gulli, M.P.; De Virgilio, C. Amino acid deprivation inhibits TORC1 through a GTPase-activating protein complex for the Rag family GTPase Gtr1. Sci. Signal. 2013, 6, ra42. [Google Scholar] [CrossRef] [Green Version]
  58. Kamada, Y.; Funakoshi, T.; Shintani, T.; Nagano, K.; Ohsumi, M.; Ohsumi, Y. Tor-mediated induction of autophagy via an Apg1 protein kinase complex. J. Cell Biol. 2000, 150, 1507–1513. [Google Scholar] [CrossRef] [Green Version]
  59. Kamada, Y.; Yoshino, K.; Kondo, C.; Kawamata, T.; Oshiro, N.; Yonezawa, K.; Ohsumi, Y. Tor directly controls the Atg1 kinase complex to regulate autophagy. Mol. Cell. Biol. 2010, 30, 1049–1058. [Google Scholar] [CrossRef] [Green Version]
  60. Tate, J.J.; Georis, I.; Dubois, E.; Cooper, T.G. Distinct phosphatase requirements and GATA factor responses to nitrogen catabolite repression and rapamycin treatment in Saccharomyces cerevisiae. J. Biol. Chem. 2010, 285, 17880–17895. [Google Scholar] [CrossRef] [Green Version]
  61. Valenzuela, L.; Aranda, C.; Gonzalez, A. TOR modulates GCN4-dependent expression of genes turned on by nitrogen limitation. J. Bacteriol. 2001, 183, 2331–2334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Bernard, A.; Jin, M.; Xu, Z.; Klionsky, D.J. A large-scale analysis of autophagy-related gene expression identifies new regulators of autophagy. Autophagy 2015, 11, 2114–2122. [Google Scholar] [CrossRef] [PubMed]
  63. Chan, T.F.; Bertram, P.G.; Ai, W.; Zheng, X.F. Regulation of APG14 expression by the GATA-type transcription factor Gln3p. J. Biol. Chem. 2001, 276, 6463–6467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yao, Z.; Delorme-Axford, E.; Backues, S.K.; Klionsky, D.J. Atg41/Icy2 regulates autophagosome formation. Autophagy 2015, 11, 2288–2299. [Google Scholar] [CrossRef] [Green Version]
  65. Swinnen, E.; Ghillebert, R.; Wilms, T.; Winderickx, J. Molecular mechanisms linking the evolutionary conserved TORC1-Sch9 nutrient signalling branch to lifespan regulation in Saccharomyces cerevisiae. FEMS Yeast Res. 2014, 14, 17–32. [Google Scholar] [CrossRef] [Green Version]
  66. Bernard, A.; Jin, M.; Gonzalez-Rodriguez, P.; Fullgrabe, J.; Delorme-Axford, E.; Backues, S.K.; Joseph, B.; Klionsky, D.J. Rph1/KDM4 mediates nutrient-limitation signaling that leads to the transcriptional induction of autophagy. Curr. Biol. 2015, 25, 546–555. [Google Scholar] [CrossRef] [Green Version]
  67. Bartholomew, C.R.; Suzuki, T.; Du, Z.; Backues, S.K.; Jin, M.; Lynch-Day, M.A.; Umekawa, M.; Kamath, A.; Zhao, M.; Xie, Z.; et al. Ume6 transcription factor is part of a signaling cascade that regulates autophagy. Proc. Natl. Acad. Sci. USA 2012, 109, 11206–11210. [Google Scholar] [CrossRef] [Green Version]
  68. Hu, G.; McQuiston, T.; Bernard, A.; Park, Y.D.; Qiu, J.; Vural, A.; Zhang, N.; Waterman, S.R.; Blewett, N.H.; Myers, T.G.; et al. A conserved mechanism of TOR-dependent RCK-mediated mRNA degradation regulates autophagy. Nat. Cell Biol. 2015, 17, 930–942. [Google Scholar] [CrossRef] [Green Version]
  69. Jin, M.; He, D.; Backues, S.K.; Freeberg, M.A.; Liu, X.; Kim, J.K.; Klionsky, D.J. Transcriptional regulation by Pho23 modulates the frequency of autophagosome formation. Curr. Biol. 2014, 24, 1314–1322. [Google Scholar] [CrossRef] [Green Version]
  70. Yu, F.; Imamura, Y.; Ueno, M.; Suzuki, S.W.; Ohsumi, Y.; Yukawa, M.; Tsuchiya, E. The yeast chromatin remodeler Rsc1-RSC complex is required for transcriptional activation of autophagy-related genes and inhibition of the TORC1 pathway in response to nitrogen starvation. Biochem. Biophys. Res. Commun. 2015, 464, 1248–1253. [Google Scholar] [CrossRef]
  71. Wen, X.; Gatica, D.; Yin, Z.; Hu, Z.; Dengjel, J.; Klionsky, D.J. The transcription factor Spt4-Spt5 complex regulates the expression of ATG8 and ATG41. Autophagy 2020, 16, 1172–1185. [Google Scholar] [CrossRef]
  72. Delorme-Axford, E.; Abernathy, E.; Lennemann, N.J.; Bernard, A.; Ariosa, A.; Coyne, C.B.; Kirkegaard, K.; Klionsky, D.J. The exoribonuclease Xrn1 is a post-transcriptional negative regulator of autophagy. Autophagy 2018, 14, 898–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Liu, X.; Yao, Z.; Jin, M.; Namkoong, S.; Yin, Z.; Lee, J.H.; Klionsky, D.J. Dhh1 promotes autophagy-related protein translation during nitrogen starvation. PLoS Biol. 2019, 17, e3000219. [Google Scholar] [CrossRef]
  74. Gatica, D.; Hu, G.; Liu, X.; Zhang, N.; Williamson, P.R.; Klionsky, D.J. The Pat1-Lsm Complex Stabilizes ATG mRNA during Nitrogen Starvation-Induced Autophagy. Mol. Cell 2019, 73, 314–324.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Yin, Z.; Liu, X.; Ariosa, A.; Huang, H.; Jin, M.; Karbstein, K.; Klionsky, D.J. Psp2, a novel regulator of autophagy that promotes autophagy-related protein translation. Cell Res. 2019, 29, 994–1008. [Google Scholar] [CrossRef]
  76. Lahiri, V.; Metur, S.P.; Hu, Z.; Song, X.; Mari, M.; Hawkins, W.D.; Bhattarai, J.; Delorme-Axford, E.; Reggiori, F.; Tang, D.; et al. Post-transcriptional regulation of ATG1 is a critical node that modulates autophagy during distinct nutrient stresses. Autophagy 2022, in press. [Google Scholar] [CrossRef] [PubMed]
  77. Tanaka, C.; Tan, L.J.; Mochida, K.; Kirisako, H.; Koizumi, M.; Asai, E.; Sakoh-Nakatogawa, M.; Ohsumi, Y.; Nakatogawa, H. Hrr25 triggers selective autophagy-related pathways by phosphorylating receptor proteins. J. Cell Biol. 2014, 207, 91–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Feng, Y.; Ariosa, A.R.; Yang, Y.; Hu, Z.; Dengjel, J.; Klionsky, D.J. Downregulation of autophagy by Met30-mediated Atg9 ubiquitination. Proc. Natl. Acad. Sci. USA 2021, 118, e2005539118. [Google Scholar] [CrossRef]
  79. Yi, C.; Ma, M.; Ran, L.; Zheng, J.; Tong, J.; Zhu, J.; Ma, C.; Sun, Y.; Zhang, S.; Feng, W.; et al. Function and molecular mechanism of acetylation in autophagy regulation. Science 2012, 336, 474–477. [Google Scholar] [CrossRef]
  80. Fullgrabe, J.; Lynch-Day, M.A.; Heldring, N.; Li, W.; Struijk, R.B.; Ma, Q.; Hermanson, O.; Rosenfeld, M.G.; Klionsky, D.J.; Joseph, B. The histone H4 lysine 16 acetyltransferase hMOF regulates the outcome of autophagy. Nature 2013, 500, 468–471. [Google Scholar] [CrossRef]
  81. Liu, X.; Jin, M.; Yao, Z.; Bernard, A.; Klionsky, D.J. Bidirectional roles of Dhh1 in regulating autophagy. Autophagy 2019, 15, 1838–1839. [Google Scholar] [CrossRef] [PubMed]
  82. Noda, T. Regulation of Autophagy through TORC1 and mTORC1. Biomolecules 2017, 7, 52. [Google Scholar] [CrossRef] [PubMed]
  83. Sullivan, A.; Wallace, R.L.; Wellington, R.; Luo, X.; Capaldi, A.P. Multilayered regulation of TORC1-body formation in budding yeast. Mol. Biol. Cell 2019, 30, 400–410. [Google Scholar] [CrossRef] [PubMed]
  84. Ukai, H.; Araki, Y.; Kira, S.; Oikawa, Y.; May, A.I.; Noda, T. Gtr/Ego-independent TORC1 activation is achieved through a glutamine-sensitive interaction with Pib2 on the vacuolar membrane. PLoS Genet. 2018, 14, e1007334. [Google Scholar] [CrossRef] [Green Version]
  85. Hatakeyama, R.; Peli-Gulli, M.P.; Hu, Z.; Jaquenoud, M.; Garcia Osuna, G.M.; Sardu, A.; Dengjel, J.; De Virgilio, C. Spatially Distinct Pools of TORC1 Balance Protein Homeostasis. Mol. Cell 2019, 73, 325–338.e8. [Google Scholar] [CrossRef] [Green Version]
  86. McDaniel, S.L.; Hepperla, A.J.; Huang, J.; Dronamraju, R.; Adams, A.T.; Kulkarni, V.G.; Davis, I.J.; Strahl, B.D. H3K36 Methylation Regulates Nutrient Stress Response in Saccharomyces cerevisiae by Enforcing Transcriptional Fidelity. Cell Rep. 2017, 19, 2371–2382. [Google Scholar] [CrossRef] [Green Version]
  87. Bar-Peled, L.; Sabatini, D.M. Regulation of mTORC1 by amino acids. Trends Cell Biol. 2014, 24, 400–406. [Google Scholar] [CrossRef]
  88. Sancak, Y.; Peterson, T.R.; Shaul, Y.D.; Lindquist, R.A.; Thoreen, C.C.; Bar-Peled, L.; Sabatini, D.M. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 2008, 320, 1496–1501. [Google Scholar] [CrossRef] [Green Version]
  89. Kim, E.; Goraksha-Hicks, P.; Li, L.; Neufeld, T.P.; Guan, K.L. Regulation of TORC1 by Rag GTPases in nutrient response. Nat. Cell Biol. 2008, 10, 935–945. [Google Scholar] [CrossRef] [Green Version]
  90. Powis, K.; De Virgilio, C. Conserved regulators of Rag GTPases orchestrate amino acid-dependent TORC1 signaling. Cell Discov. 2016, 2, 15049. [Google Scholar] [CrossRef] [Green Version]
  91. Zoncu, R.; Bar-Peled, L.; Efeyan, A.; Wang, S.; Sancak, Y.; Sabatini, D.M. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase. Science 2011, 334, 678–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bar-Peled, L.; Schweitzer, L.D.; Zoncu, R.; Sabatini, D.M. Ragulator is a GEF for the rag GTPases that signal amino acid levels to mTORC1. Cell 2012, 150, 1196–1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Shen, K.; Choe, A.; Sabatini, D.M. Intersubunit Crosstalk in the Rag GTPase Heterodimer Enables mTORC1 to Respond Rapidly to Amino Acid Availability. Mol. Cell 2017, 68, 552–565. [Google Scholar] [CrossRef] [PubMed]
  94. Bar-Peled, L.; Chantranupong, L.; Cherniack, A.D.; Chen, W.W.; Ottina, K.A.; Grabiner, B.C.; Spear, E.D.; Carter, S.L.; Meyerson, M.; Sabatini, D.M. A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 2013, 340, 1100–1106. [Google Scholar] [CrossRef] [Green Version]
  95. Menon, S.; Dibble, C.C.; Talbott, G.; Hoxhaj, G.; Valvezan, A.J.; Takahashi, H.; Cantley, L.C.; Manning, B.D. Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome. Cell 2014, 156, 771–785. [Google Scholar] [CrossRef] [Green Version]
  96. Demetriades, C.; Plescher, M.; Teleman, A.A. Lysosomal recruitment of TSC2 is a universal response to cellular stress. Nat. Commun. 2016, 7, 10662. [Google Scholar] [CrossRef]
  97. Yao, Y.; Hong, S.; Ikeda, T.; Mori, H.; MacDougald, O.A.; Nada, S.; Okada, M.; Inoki, K. Amino Acids Enhance Polyubiquitination of Rheb and Its Binding to mTORC1 by Blocking Lysosomal ATXN3 Deubiquitinase Activity. Mol. Cell 2020, 80, 437–451.e6. [Google Scholar] [CrossRef]
  98. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [Green Version]
  99. Hosokawa, N.; Hara, T.; Kaizuka, T.; Kishi, C.; Takamura, A.; Miura, Y.; Iemura, S.; Natsume, T.; Takehana, K.; Yamada, N.; et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol. Biol. Cell 2009, 20, 1981–1991. [Google Scholar] [CrossRef] [Green Version]
  100. Chan, E.Y. mTORC1 phosphorylates the ULK1-mAtg13-FIP200 autophagy regulatory complex. Sci. Signal. 2009, 2, pe51. [Google Scholar] [CrossRef]
  101. Dunlop, E.A.; Hunt, D.K.; Acosta-Jaquez, H.A.; Fingar, D.C.; Tee, A.R. ULK1 inhibits mTORC1 signaling, promotes multisite Raptor phosphorylation and hinders substrate binding. Autophagy 2011, 7, 737–747. [Google Scholar] [CrossRef] [PubMed]
  102. Jung, C.H.; Seo, M.; Otto, N.M.; Kim, D.H. ULK1 inhibits the kinase activity of mTORC1 and cell proliferation. Autophagy 2011, 7, 1212–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yuan, H.X.; Russell, R.C.; Guan, K.L. Regulation of PIK3C3/VPS34 complexes by MTOR in nutrient stress-induced autophagy. Autophagy 2013, 9, 1983–1995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ma, X.; Zhang, S.; He, L.; Rong, Y.; Brier, L.W.; Sun, Q.; Liu, R.; Fan, W.; Chen, S.; Yue, Z.; et al. MTORC1-mediated NRBF2 phosphorylation functions as a switch for the class III PtdIns3K and autophagy. Autophagy 2017, 13, 592–607. [Google Scholar] [CrossRef] [Green Version]
  105. Nazio, F.; Strappazzon, F.; Antonioli, M.; Bielli, P.; Cianfanelli, V.; Bordi, M.; Gretzmeier, C.; Dengjel, J.; Piacentini, M.; Fimia, G.M.; et al. mTOR inhibits autophagy by controlling ULK1 ubiquitylation, self-association and function through AMBRA1 and TRAF6. Nat. Cell Biol. 2013, 15, 406–416. [Google Scholar] [CrossRef] [PubMed]
  106. Kim, Y.M.; Jung, C.H.; Seo, M.; Kim, E.K.; Park, J.M.; Bae, S.S.; Kim, D.H. mTORC1 phosphorylates UVRAG to negatively regulate autophagosome and endosome maturation. Mol. Cell 2015, 57, 207–218. [Google Scholar] [CrossRef] [Green Version]
  107. Wan, W.; You, Z.; Zhou, L.; Xu, Y.; Peng, C.; Zhou, T.; Yi, C.; Shi, Y.; Liu, W. mTORC1-Regulated and HUWE1-Mediated WIPI2 Degradation Controls Autophagy Flux. Mol. Cell 2018, 72, 303–315.e6. [Google Scholar] [CrossRef] [Green Version]
  108. Wan, W.; You, Z.; Xu, Y.; Zhou, L.; Guan, Z.; Peng, C.; Wong, C.C.L.; Su, H.; Zhou, T.; Xia, H.; et al. mTORC1 Phosphorylates Acetyltransferase p300 to Regulate Autophagy and Lipogenesis. Mol. Cell 2017, 68, 323–335. [Google Scholar] [CrossRef] [Green Version]
  109. Lee, I.H.; Finkel, T. Regulation of autophagy by the p300 acetyltransferase. J. Biol. Chem. 2009, 284, 6322–6328. [Google Scholar] [CrossRef] [Green Version]
  110. Napolitano, G.; Ballabio, A. TFEB at a glance. J. Cell Sci. 2016, 129, 2475–2481. [Google Scholar] [CrossRef] [Green Version]
  111. Martina, J.A.; Puertollano, R. Rag GTPases mediate amino acid-dependent recruitment of TFEB and MITF to lysosomes. J. Cell Biol. 2013, 200, 475–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Settembre, C.; Di Malta, C.; Polito, V.A.; Garcia Arencibia, M.; Vetrini, F.; Erdin, S.; Erdin, S.U.; Huynh, T.; Medina, D.; Colella, P.; et al. TFEB links autophagy to lysosomal biogenesis. Science 2011, 332, 1429–1433. [Google Scholar] [CrossRef] [Green Version]
  113. Roczniak-Ferguson, A.; Petit, C.S.; Froehlich, F.; Qian, S.; Ky, J.; Angarola, B.; Walther, T.C.; Ferguson, S.M. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci. Signal. 2012, 5, ra42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Martina, J.A.; Chen, Y.; Gucek, M.; Puertollano, R. MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy 2012, 8, 903–914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Vega-Rubin-de-Celis, S.; Pena-Llopis, S.; Konda, M.; Brugarolas, J. Multistep regulation of TFEB by MTORC1. Autophagy 2017, 13, 464–472. [Google Scholar] [CrossRef] [Green Version]
  116. Napolitano, G.; Esposito, A.; Choi, H.; Matarese, M.; Benedetti, V.; Di Malta, C.; Monfregola, J.; Medina, D.L.; Lippincott-Schwartz, J.; Ballabio, A. mTOR-dependent phosphorylation controls TFEB nuclear export. Nat. Commun. 2018, 9, 3312. [Google Scholar] [CrossRef]
  117. Wang, Y.; Huang, Y.; Liu, J.; Zhang, J.; Xu, M.; You, Z.; Peng, C.; Gong, Z.; Liu, W. Acetyltransferase GCN5 regulates autophagy and lysosome biogenesis by targeting TFEB. EMBO Rep. 2020, 21, e48335. [Google Scholar] [CrossRef] [PubMed]
  118. Martina, J.A.; Diab, H.I.; Lishu, L.; Jeong, A.L.; Patange, S.; Raben, N.; Puertollano, R. The nutrient-responsive transcription factor TFE3 promotes autophagy, lysosomal biogenesis, and clearance of cellular debris. Sci. Signal. 2014, 7, ra9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Ploper, D.; Taelman, V.F.; Robert, L.; Perez, B.S.; Titz, B.; Chen, H.W.; Graeber, T.G.; von Euw, E.; Ribas, A.; De Robertis, E.M. MITF drives endolysosomal biogenesis and potentiates Wnt signaling in melanoma cells. Proc. Natl. Acad. Sci. USA 2015, 112, E420–E429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Moller, K.; Sigurbjornsdottir, S.; Arnthorsson, A.O.; Pogenberg, V.; Dilshat, R.; Fock, V.; Brynjolfsdottir, S.H.; Bindesboll, C.; Bessadottir, M.; Ogmundsdottir, H.M.; et al. MITF has a central role in regulating starvation-induced autophagy in melanoma. Sci. Rep. 2019, 9, 1055. [Google Scholar] [CrossRef] [Green Version]
  121. Di Malta, C.; Siciliano, D.; Calcagni, A.; Monfregola, J.; Punzi, S.; Pastore, N.; Eastes, A.N.; Davis, O.; De Cegli, R.; Zampelli, A.; et al. Transcriptional activation of RagD GTPase controls mTORC1 and promotes cancer growth. Science 2017, 356, 1188–1192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Wengrod, J.; Wang, D.; Weiss, S.; Zhong, H.; Osman, I.; Gardner, L.B. Phosphorylation of eIF2alpha triggered by mTORC1 inhibition and PP6C activation is required for autophagy and is aberrant in PP6C-mutated melanoma. Sci. Signal. 2015, 8, ra27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. B’Chir, W.; Maurin, A.C.; Carraro, V.; Averous, J.; Jousse, C.; Muranishi, Y.; Parry, L.; Stepien, G.; Fafournoux, P.; Bruhat, A. The eIF2alpha/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Res. 2013, 41, 7683–7699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Tang, H.W.; Weng, J.H.; Lee, W.X.; Hu, Y.; Gu, L.; Cho, S.; Lee, G.; Binari, R.; Li, C.; Cheng, M.E.; et al. mTORC1-chaperonin CCT signaling regulates m(6)A RNA methylation to suppress autophagy. Proc. Natl. Acad. Sci. USA 2021, 118, e2021945118. [Google Scholar] [CrossRef] [PubMed]
  125. Baek, S.H.; Kim, K.I. Epigenetic Control of Autophagy: Nuclear Events Gain More Attention. Mol. Cell 2017, 65, 781–785. [Google Scholar] [CrossRef] [Green Version]
  126. Bowman, C.J.; Ayer, D.E.; Dynlacht, B.D. Foxk proteins repress the initiation of starvation-induced atrophy and autophagy programs. Nat. Cell Biol. 2014, 16, 1202–1214. [Google Scholar] [CrossRef] [PubMed]
  127. Wei, Y.; Pattingre, S.; Sinha, S.; Bassik, M.; Levine, B. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol. Cell 2008, 30, 678–688. [Google Scholar] [CrossRef] [Green Version]
  128. Wei, Y.; An, Z.; Zou, Z.; Sumpter, R.; Su, M.; Zang, X.; Sinha, S.; Gaestel, M.; Levine, B. The stress-responsive kinases MAPKAPK2/MAPKAPK3 activate starvation-induced autophagy through Beclin 1 phosphorylation. Elife 2015, 4, e05289. [Google Scholar] [CrossRef]
  129. Comb, W.C.; Cogswell, P.; Sitcheran, R.; Baldwin, A.S. IKK-dependent, NF-kappaB-independent control of autophagic gene expression. Oncogene 2011, 30, 1727–1732. [Google Scholar] [CrossRef] [Green Version]
  130. Hardie, D.G.; Carling, D.; Halford, N. Roles of the Snf1/Rkin1/AMP-activated protein kinase family in the response to environmental and nutritional stress. Semin. Cell Biol. 1994, 5, 409–416. [Google Scholar] [CrossRef]
  131. Herzig, S.; Shaw, R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 121–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Hawley, S.A.; Davison, M.; Woods, A.; Davies, S.P.; Beri, R.K.; Carling, D.; Hardie, D.G. Characterization of the AMP-activated protein kinase kinase from rat liver and identification of threonine 172 as the major site at which it phosphorylates AMP-activated protein kinase. J. Biol. Chem. 1996, 271, 27879–27887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Xiao, B.; Heath, R.; Saiu, P.; Leiper, F.C.; Leone, P.; Jing, C.; Walker, P.A.; Haire, L.; Eccleston, J.F.; Davis, C.T.; et al. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature 2007, 449, 496–500. [Google Scholar] [CrossRef] [PubMed]
  134. Hardie, D.G.; Scott, J.W.; Pan, D.A.; Hudson, E.R. Management of cellular energy by the AMP-activated protein kinase system. FEBS Lett. 2003, 546, 113–120. [Google Scholar] [CrossRef]
  135. Hawley, S.A.; Selbert, M.A.; Goldstein, E.G.; Edelman, A.M.; Carling, D.; Hardie, D.G. 5′-AMP activates the AMP-activated protein kinase cascade, and Ca2+/calmodulin activates the calmodulin-dependent protein kinase I cascade, via three independent mechanisms. J. Biol. Chem. 1995, 270, 27186–27191. [Google Scholar] [CrossRef] [Green Version]
  136. Shaw, R.J.; Kosmatka, M.; Bardeesy, N.; Hurley, R.L.; Witters, L.A.; DePinho, R.A.; Cantley, L.C. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc. Natl. Acad. Sci. USA 2004, 101, 3329–3335. [Google Scholar] [CrossRef] [Green Version]
  137. Davies, S.P.; Helps, N.R.; Cohen, P.T.; Hardie, D.G. 5′-AMP inhibits dephosphorylation, as well as promoting phosphorylation, of the AMP-activated protein kinase. Studies using bacterially expressed human protein phosphatase-2C alpha and native bovine protein phosphatase-2AC. FEBS Lett. 1995, 377, 421–425. [Google Scholar] [CrossRef] [Green Version]
  138. Gowans, G.J.; Hawley, S.A.; Ross, F.A.; Hardie, D.G. AMP is a true physiological regulator of AMP-activated protein kinase by both allosteric activation and enhancing net phosphorylation. Cell Metab. 2013, 18, 556–566. [Google Scholar] [CrossRef] [Green Version]
  139. Gwinn, D.M.; Shackelford, D.B.; Egan, D.F.; Mihaylova, M.M.; Mery, A.; Vasquez, D.S.; Turk, B.E.; Shaw, R.J. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 2008, 30, 214–226. [Google Scholar] [CrossRef] [Green Version]
  140. Inoki, K.; Zhu, T.; Guan, K.L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 2003, 115, 577–590. [Google Scholar] [CrossRef] [Green Version]
  141. Inoki, K.; Li, Y.; Xu, T.; Guan, K.L. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 2003, 17, 1829–1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Russell, R.C.; Tian, Y.; Yuan, H.; Park, H.W.; Chang, Y.Y.; Kim, J.; Kim, H.; Neufeld, T.P.; Dillin, A.; Guan, K.L. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat. Cell Biol. 2013, 15, 741–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Weerasekara, V.K.; Panek, D.J.; Broadbent, D.G.; Mortenson, J.B.; Mathis, A.D.; Logan, G.N.; Prince, J.T.; Thomson, D.M.; Thompson, J.W.; Andersen, J.L. Metabolic-stress-induced rearrangement of the 14-3-3zeta interactome promotes autophagy via a ULK1- and AMPK-regulated 14-3-3zeta interaction with phosphorylated Atg9. Mol. Cell. Biol. 2014, 34, 4379–4388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Greer, E.L.; Oskoui, P.R.; Banko, M.R.; Maniar, J.M.; Gygi, M.P.; Gygi, S.P.; Brunet, A. The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J. Biol. Chem. 2007, 282, 30107–30119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Tamargo-Gomez, I.; Marino, G. AMPK: Regulation of Metabolic Dynamics in the Context of Autophagy. Int. J. Mol. Sci. 2018, 19, 3812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Chang, C.; Su, H.; Zhang, D.; Wang, Y.; Shen, Q.; Liu, B.; Huang, R.; Zhou, T.; Peng, C.; Wong, C.C.; et al. AMPK-Dependent Phosphorylation of GAPDH Triggers Sirt1 Activation and Is Necessary for Autophagy upon Glucose Starvation. Mol. Cell 2015, 60, 930–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Lee, I.H.; Cao, L.; Mostoslavsky, R.; Lombard, D.B.; Liu, J.; Bruns, N.E.; Tsokos, M.; Alt, F.W.; Finkel, T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc. Natl. Acad. Sci. USA 2008, 105, 3374–3379. [Google Scholar] [CrossRef] [Green Version]
  148. Hariharan, N.; Maejima, Y.; Nakae, J.; Paik, J.; Depinho, R.A.; Sadoshima, J. Deacetylation of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy in Cardiac Myocytes. Circ. Res. 2010, 107, 1470–1482. [Google Scholar] [CrossRef] [Green Version]
  149. Hu, Y.; Chen, H.; Zhang, L.; Lin, X.; Li, X.; Zhuang, H.; Fan, H.; Meng, T.; He, Z.; Huang, H.; et al. The AMPK-MFN2 axis regulates MAM dynamics and autophagy induced by energy stresses. Autophagy 2021, 17, 1142–1156. [Google Scholar] [CrossRef]
  150. Finkel, T. Signal transduction by reactive oxygen species. J. Cell Biol. 2011, 194, 7–15. [Google Scholar] [CrossRef] [Green Version]
  151. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef] [PubMed]
  152. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gao, Q. Oxidative Stress and Autophagy. Adv. Exp. Med. Biol. 2019, 1206, 179–198. [Google Scholar] [CrossRef] [PubMed]
  154. Filomeni, G.; De Zio, D.; Cecconi, F. Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 2015, 22, 377–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kiffin, R.; Bandyopadhyay, U.; Cuervo, A.M. Oxidative stress and autophagy. Antioxid. Redox Signal. 2006, 8, 152–162. [Google Scholar] [CrossRef] [PubMed]
  156. Chen, Y.; McMillan-Ward, E.; Kong, J.; Israels, S.J.; Gibson, S.B. Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death Differ. 2008, 15, 171–182. [Google Scholar] [CrossRef]
  157. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428. [Google Scholar] [CrossRef]
  158. Dobrowolny, G.; Aucello, M.; Rizzuto, E.; Beccafico, S.; Mammucari, C.; Boncompagni, S.; Belia, S.; Wannenes, F.; Nicoletti, C.; Del Prete, Z.; et al. Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. Cell Metab. 2008, 8, 425–436. [Google Scholar] [CrossRef]
  159. Park, E.; Chung, S.W. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019, 10, 822. [Google Scholar] [CrossRef] [Green Version]
  160. Scherz-Shouval, R.; Elazar, Z. Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem. Sci. 2011, 36, 30–38. [Google Scholar] [CrossRef]
  161. Scherz-Shouval, R.; Elazar, Z. ROS, mitochondria and the regulation of autophagy. Trends Cell Biol. 2007, 17, 422–427. [Google Scholar] [CrossRef] [PubMed]
  162. Lee, J.; Giordano, S.; Zhang, J. Autophagy, mitochondria and oxidative stress: Cross-talk and redox signalling. Biochem. J. 2012, 441, 523–540. [Google Scholar] [CrossRef] [Green Version]
  163. Hayyan, M.; Hashim, M.A.; AlNashef, I.M. Superoxide Ion: Generation and Chemical Implications. Chem. Rev. 2016, 116, 3029–3085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Groeger, G.; Quiney, C.; Cotter, T.G. Hydrogen peroxide as a cell-survival signaling molecule. Antioxid. Redox Signal. 2009, 11, 2655–2671. [Google Scholar] [CrossRef]
  165. Marinho, H.S.; Real, C.; Cyrne, L.; Soares, H.; Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014, 2, 535–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Lloyd, R.V.; Hanna, P.M.; Mason, R.P. The origin of the hydroxyl radical oxygen in the Fenton reaction. Free Radic. Biol. Med. 1997, 22, 885–888. [Google Scholar] [CrossRef]
  167. Halliwell, B.; Chirico, S. Lipid peroxidation: Its mechanism, measurement, and significance. Am. J. Clin. Nutr. 1993, 57, 715S–724S, discussion 724S—725S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Zhang, H.; Kong, X.; Kang, J.; Su, J.; Li, Y.; Zhong, J.; Sun, L. Oxidative stress induces parallel autophagy and mitochondria dysfunction in human glioma U251 cells. Toxicol. Sci. 2009, 110, 376–388. [Google Scholar] [CrossRef]
  169. Chen, Y.; Azad, M.B.; Gibson, S.B. Superoxide is the major reactive oxygen species regulating autophagy. Cell Death Differ. 2009, 16, 1040–1052. [Google Scholar] [CrossRef] [Green Version]
  170. Kim, E.H.; Sohn, S.; Kwon, H.J.; Kim, S.U.; Kim, M.J.; Lee, S.J.; Choi, K.S. Sodium selenite induces superoxide-mediated mitochondrial damage and subsequent autophagic cell death in malignant glioma cells. Cancer Res. 2007, 67, 6314–6324. [Google Scholar] [CrossRef] [Green Version]
  171. Chen, J.W.; Ni, B.B.; Li, B.; Yang, Y.H.; Jiang, S.D.; Jiang, L.S. The responses of autophagy and apoptosis to oxidative stress in nucleus pulposus cells: Implications for disc degeneration. Cell. Physiol. Biochem. 2014, 34, 1175–1189. [Google Scholar] [CrossRef] [PubMed]
  172. Yu, Y.; Fan, S.M.; Song, J.K.; Tashiro, S.; Onodera, S.; Ikejima, T. Hydroxyl radical (·OH) played a pivotal role in oridonin-induced apoptosis and autophagy in human epidermoid carcinoma A431 cells. Biol. Pharm. Bull. 2012, 35, 2148–2159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Barsoum, M.J.; Yuan, H.; Gerencser, A.A.; Liot, G.; Kushnareva, Y.; Gräber, S.; Kovacs, I.; Lee, W.D.; Waggoner, J.; Cui, J.; et al. Nitric oxide-induced mitochondrial fission is regulated by dynamin-related GTPases in neurons. EMBO J. 2006, 25, 3900–3911. [Google Scholar] [CrossRef] [PubMed]
  174. Liot, G.; Bossy, B.; Lubitz, S.; Kushnareva, Y.; Sejbuk, N.; Bossy-Wetzel, E. Complex II inhibition by 3-NP causes mitochondrial fragmentation and neuronal cell death via an NMDA- and ROS-dependent pathway. Cell Death Differ. 2009, 16, 899–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Tyagi, N.; Vacek, J.C.; Givvimani, S.; Sen, U.; Tyagi, S.C. Cardiac specific deletion of N-methyl-d-aspartate receptor 1 ameliorates mtMMP-9 mediated autophagy/mitophagy in hyperhomocysteinemia. J. Recept. Signal Transduct. Res. 2010, 30, 78–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Dodson, M.; Wani, W.Y.; Redmann, M.; Benavides, G.A.; Johnson, M.S.; Ouyang, X.; Cofield, S.S.; Mitra, K.; Darley-Usmar, V.; Zhang, J. Regulation of autophagy, mitochondrial dynamics, and cellular bioenergetics by 4-hydroxynonenal in primary neurons. Autophagy 2017, 13, 1828–1840. [Google Scholar] [CrossRef] [Green Version]
  177. De Gaetano, A.; Gibellini, L.; Zanini, G.; Nasi, M.; Cossarizza, A.; Pinti, M. Mitophagy and Oxidative Stress: The Role of Aging. Antioxidants 2021, 10, 794. [Google Scholar] [CrossRef]
  178. Deffieu, M.; Bhatia-Kissová, I.; Salin, B.; Galinier, A.; Manon, S.; Camougrand, N. Glutathione participates in the regulation of mitophagy in yeast. J. Biol. Chem. 2009, 284, 14828–14837. [Google Scholar] [CrossRef] [Green Version]
  179. Jing, H.; Liu, H.; Zhang, L.; Gao, J.; Song, H.; Tan, X. Ethanol Induces Autophagy Regulated by Mitochondrial ROS in Saccharomyces cerevisiae. J. Microbiol. Biotechnol. 2018, 28, 1982–1991. [Google Scholar] [CrossRef] [Green Version]
  180. Rodrigues-Pousada, C.; Devaux, F.; Caetano, S.M.; Pimentel, C.; da Silva, S.; Cordeiro, A.C.; Amaral, C. Yeast AP-1 like transcription factors (Yap) and stress response: A current overview. Microb. Cell 2019, 6, 267–285. [Google Scholar] [CrossRef]
  181. Toledano, M.B.; Delaunay, A.; Monceau, L.; Tacnet, F. Microbial H2O2 sensors as archetypical redox signaling modules. Trends Biochem. Sci. 2004, 29, 351–357. [Google Scholar] [CrossRef] [PubMed]
  182. Harshman, K.D.; Moye-Rowley, W.S.; Parker, C.S. Transcriptional activation by the SV40 AP-1 recognition element in yeast is mediated by a factor similar to AP-1 that is distinct from GCN4. Cell 1988, 53, 321–330. [Google Scholar] [CrossRef]
  183. Fernandes, L.; Rodrigues-Pousada, C.; Struhl, K. Yap, a novel family of eight bZIP proteins in Saccharomyces cerevisiae with distinct biological functions. Mol. Cell. Biol. 1997, 17, 6982–6993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Kuge, S.; Jones, N. YAP1 dependent activation of TRX2 is essential for the response of Saccharomyces cerevisiae to oxidative stress by hydroperoxides. EMBO J. 1994, 13, 655–664. [Google Scholar] [CrossRef]
  185. Kuge, S.; Jones, N.; Nomoto, A. Regulation of yAP-1 nuclear localization in response to oxidative stress. EMBO J. 1997, 16, 1710–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Delaunay, A.; Isnard, A.D.; Toledano, M.B. H2O2 sensing through oxidation of the Yap1 transcription factor. EMBO J. 2000, 19, 5157–5166. [Google Scholar] [CrossRef] [PubMed]
  187. Okazaki, S.; Tachibana, T.; Naganuma, A.; Mano, N.; Kuge, S. Multistep disulfide bond formation in Yap1 is required for sensing and transduction of H2O2 stress signal. Mol. Cell 2007, 27, 675–688. [Google Scholar] [CrossRef]
  188. Delaunay, A.; Pflieger, D.; Barrault, M.B.; Vinh, J.; Toledano, M.B. A thiol peroxidase is an H2O2 receptor and redox-transducer in gene activation. Cell 2002, 111, 471–481. [Google Scholar] [CrossRef] [Green Version]
  189. Maeda, Y.; Oku, M.; Sakai, Y. A defect of the vacuolar putative lipase Atg15 accelerates degradation of lipid droplets through lipolysis. Autophagy 2015, 11, 1247–1258. [Google Scholar] [CrossRef] [Green Version]
  190. Ramya, V.; Rajasekharan, R. ATG15 encodes a phospholipase and is transcriptionally regulated by YAP1 in Saccharomyces cerevisiae. FEBS Lett. 2016, 590, 3155–3167. [Google Scholar] [CrossRef]
  191. Scherz-Shouval, R.; Shvets, E.; Fass, E.; Shorer, H.; Gil, L.; Elazar, Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 2007, 26, 1749–1760. [Google Scholar] [CrossRef] [PubMed]
  192. Pérez-Pérez, M.E.; Zaffagnini, M.; Marchand, C.H.; Crespo, J.L.; Lemaire, S.D. The yeast autophagy protease Atg4 is regulated by thioredoxin. Autophagy 2014, 10, 1953–1964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Scherz-Shouval, R.; Elazar, Z. Monitoring starvation-induced reactive oxygen species formation. Methods Enzymol. 2009, 452, 119–130. [Google Scholar] [CrossRef]
  194. Mungai, P.T.; Waypa, G.B.; Jairaman, A.; Prakriya, M.; Dokic, D.; Ball, M.K.; Schumacker, P.T. Hypoxia triggers AMPK activation through reactive oxygen species-mediated activation of calcium release-activated calcium channels. Mol. Cell. Biol. 2011, 31, 3531–3545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Djavaheri-Mergny, M.; Amelotti, M.; Mathieu, J.; Besançon, F.; Bauvy, C.; Souquère, S.; Pierron, G.; Codogno, P. NF-kappaB activation represses tumor necrosis factor-alpha-induced autophagy. J. Biol. Chem. 2006, 281, 30373–30382. [Google Scholar] [CrossRef] [Green Version]
  196. Djavaheri-Mergny, M.; Javelaud, D.; Wietzerbin, J.; Besançon, F. NF-kappaB activation prevents apoptotic oxidative stress via an increase of both thioredoxin and MnSOD levels in TNFalpha-treated Ewing sarcoma cells. FEBS Lett. 2004, 578, 111–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Zhang, H.; Bosch-Marce, M.; Shimoda, L.A.; Tan, Y.S.; Baek, J.H.; Wesley, J.B.; Gonzalez, F.J.; Semenza, G.L. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J. Biol. Chem. 2008, 283, 10892–10903. [Google Scholar] [CrossRef] [Green Version]
  198. Zmijewski, J.W.; Banerjee, S.; Bae, H.; Friggeri, A.; Lazarowski, E.R.; Abraham, E. Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated protein kinase. J. Biol. Chem. 2010, 285, 33154–33164. [Google Scholar] [CrossRef] [Green Version]
  199. Bellot, G.; Garcia-Medina, R.; Gounon, P.; Chiche, J.; Roux, D.; Pouysségur, J.; Mazure, N.M. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol. Cell. Biol. 2009, 29, 2570–2581. [Google Scholar] [CrossRef] [Green Version]
  200. Alexander, A.; Kim, J.; Walker, C.L. ATM engages the TSC2/mTORC1 signaling node to regulate autophagy. Autophagy 2010, 6, 672–673. [Google Scholar] [CrossRef] [Green Version]
  201. Cirotti, C.; Rizza, S.; Giglio, P.; Poerio, N.; Allega, M.F.; Claps, G.; Pecorari, C.; Lee, J.H.; Benassi, B.; Barilà, D.; et al. Redox activation of ATM enhances GSNOR translation to sustain mitophagy and tolerance to oxidative stress. EMBO Rep. 2021, 22, e50500. [Google Scholar] [CrossRef] [PubMed]
  202. Dames, S.A.; Mulet, J.M.; Rathgeb-Szabo, K.; Hall, M.N.; Grzesiek, S. The solution structure of the FATC domain of the protein kinase target of rapamycin suggests a role for redox-dependent structural and cellular stability. J. Biol. Chem. 2005, 280, 20558–20564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Bensaad, K.; Cheung, E.C.; Vousden, K.H. Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO J. 2009, 28, 3015–3026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. McClung, J.M.; Judge, A.R.; Powers, S.K.; Yan, Z. p38 MAPK links oxidative stress to autophagy-related gene expression in cachectic muscle wasting. Am. J. Physiol. Cell Physiol. 2010, 298, C542–C549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Pajares, M.; Cuadrado, A.; Engedal, N.; Jirsova, Z.; Cahova, M. The Role of Free Radicals in Autophagy Regulation: Implications for Ageing. Oxid. Med. Cell. Longev. 2018, 2018, 2450748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Madden, S.K.; Itzhaki, L.S. Structural and mechanistic insights into the Keap1-Nrf2 system as a route to drug discovery. Biochim. Biophys. Acta Proteins Proteom. 2020, 1868, 140405. [Google Scholar] [CrossRef]
  208. Jiang, T.; Harder, B.; Rojo de la Vega, M.; Wong, P.K.; Chapman, E.; Zhang, D.D. p62 links autophagy and Nrf2 signaling. Free Radic. Biol. Med. 2015, 88, 199–204. [Google Scholar] [CrossRef] [Green Version]
  209. Fuse, Y.; Kobayashi, M. Conservation of the Keap1-Nrf2 System: An Evolutionary Journey through Stressful Space and Time. Molecules 2017, 22, 436. [Google Scholar] [CrossRef]
  210. Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997, 236, 313–322. [Google Scholar] [CrossRef]
  211. Kobayashi, A.; Kang, M.I.; Okawa, H.; Ohtsuji, M.; Zenke, Y.; Chiba, T.; Igarashi, K.; Yamamoto, M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell. Biol. 2004, 24, 7130–7139. [Google Scholar] [CrossRef] [Green Version]
  212. Zhang, D.D.; Hannink, M. Distinct cysteine residues in Keap1 are required for Keap1-dependent ubiquitination of Nrf2 and for stabilization of Nrf2 by chemopreventive agents and oxidative stress. Mol. Cell. Biol. 2003, 23, 8137–8151. [Google Scholar] [CrossRef] [Green Version]
  213. Zhang, D.D.; Lo, S.C.; Cross, J.V.; Templeton, D.J.; Hannink, M. Keap1 is a redox-regulated substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex. Mol. Cell. Biol. 2004, 24, 10941–10953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Itoh, K.; Wakabayashi, N.; Katoh, Y.; Ishii, T.; Igarashi, K.; Engel, J.D.; Yamamoto, M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999, 13, 76–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Lo, S.C.; Li, X.; Henzl, M.T.; Beamer, L.J.; Hannink, M. Structure of the Keap1:Nrf2 interface provides mechanistic insight into Nrf2 signaling. EMBO J. 2006, 25, 3605–3617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Furukawa, M.; Xiong, Y. BTB protein Keap1 targets antioxidant transcription factor Nrf2 for ubiquitination by the Cullin 3-Roc1 ligase. Mol. Cell. Biol. 2005, 25, 162–171. [Google Scholar] [CrossRef] [Green Version]
  217. Suzuki, T.; Muramatsu, A.; Saito, R.; Iso, T.; Shibata, T.; Kuwata, K.; Kawaguchi, S.I.; Iwawaki, T.; Adachi, S.; Suda, H.; et al. Molecular Mechanism of Cellular Oxidative Stress Sensing by Keap1. Cell Rep. 2019, 28, 746–758.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Hourihan, J.M.; Kenna, J.G.; Hayes, J.D. The gasotransmitter hydrogen sulfide induces nrf2-target genes by inactivating the keap1 ubiquitin ligase substrate adaptor through formation of a disulfide bond between cys-226 and cys-613. Antioxid. Redox Signal. 2013, 19, 465–481. [Google Scholar] [CrossRef]
  219. Fourquet, S.; Guerois, R.; Biard, D.; Toledano, M.B. Activation of NRF2 by nitrosative agents and H2O2 involves KEAP1 disulfide formation. J. Biol. Chem. 2010, 285, 8463–8471. [Google Scholar] [CrossRef] [Green Version]
  220. Komatsu, M.; Waguri, S.; Koike, M.; Sou, Y.S.; Ueno, T.; Hara, T.; Mizushima, N.; Iwata, J.; Ezaki, J.; Murata, S.; et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 2007, 131, 1149–1163. [Google Scholar] [CrossRef] [Green Version]
  221. Katsuragi, Y.; Ichimura, Y.; Komatsu, M. p62/SQSTM1 functions as a signaling hub and an autophagy adaptor. FEBS J. 2015, 282, 4672–4678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Jain, A.; Lamark, T.; Sjøttem, E.; Larsen, K.B.; Awuh, J.A.; Øvervatn, A.; McMahon, M.; Hayes, J.D.; Johansen, T. p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J. Biol. Chem. 2010, 285, 22576–22591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Copple, I.M.; Lister, A.; Obeng, A.D.; Kitteringham, N.R.; Jenkins, R.E.; Layfield, R.; Foster, B.J.; Goldring, C.E.; Park, B.K. Physical and functional interaction of sequestosome 1 with Keap1 regulates the Keap1-Nrf2 cell defense pathway. J. Biol. Chem. 2010, 285, 16782–16788. [Google Scholar] [CrossRef] [Green Version]
  224. Fan, W.; Tang, Z.; Chen, D.; Moughon, D.; Ding, X.; Chen, S.; Zhu, M.; Zhong, Q. Keap1 facilitates p62-mediated ubiquitin aggregate clearance via autophagy. Autophagy 2010, 6, 614–621. [Google Scholar] [CrossRef] [Green Version]
  225. Komatsu, M.; Kurokawa, H.; Waguri, S.; Taguchi, K.; Kobayashi, A.; Ichimura, Y.; Sou, Y.S.; Ueno, I.; Sakamoto, A.; Tong, K.I.; et al. The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nat. Cell Biol. 2010, 12, 213–223. [Google Scholar] [CrossRef] [PubMed]
  226. Lau, A.; Wang, X.J.; Zhao, F.; Villeneuve, N.F.; Wu, T.; Jiang, T.; Sun, Z.; White, E.; Zhang, D.D. A noncanonical mechanism of Nrf2 activation by autophagy deficiency: Direct interaction between Keap1 and p62. Mol. Cell. Biol. 2010, 30, 3275–3285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Pajares, M.; Jiménez-Moreno, N.; García-Yagüe, Á.J.; Escoll, M.; de Ceballos, M.L.; Van Leuven, F.; Rábano, A.; Yamamoto, M.; Rojo, A.I.; Cuadrado, A. Transcription factor NFE2L2/NRF2 is a regulator of macroautophagy genes. Autophagy 2016, 12, 1902–1916. [Google Scholar] [CrossRef] [Green Version]
  228. Pajares, M.; Rojo, A.I.; Arias, E.; Díaz-Carretero, A.; Cuervo, A.M.; Cuadrado, A. Transcription factor NFE2L2/NRF2 modulates chaperone-mediated autophagy through the regulation of LAMP2A. Autophagy 2018, 14, 1310–1322. [Google Scholar] [CrossRef]
  229. Meng, F.; Yao, D.; Shi, Y.; Kabakoff, J.; Wu, W.; Reicher, J.; Ma, Y.; Moosmann, B.; Masliah, E.; Lipton, S.A.; et al. Oxidation of the cysteine-rich regions of parkin perturbs its E3 ligase activity and contributes to protein aggregation. Mol. Neurodegener. 2011, 6, 34. [Google Scholar] [CrossRef] [Green Version]
  230. Vandiver, M.S.; Paul, B.D.; Xu, R.; Karuppagounder, S.; Rao, F.; Snowman, A.M.; Ko, H.S.; Lee, Y.I.; Dawson, V.L.; Dawson, T.M.; et al. Sulfhydration mediates neuroprotective actions of parkin. Nat. Commun. 2013, 4, 1626. [Google Scholar] [CrossRef] [Green Version]
  231. Astuti, R.I.; Nasuno, R.; Takagi, H. Nitric oxide signaling in yeast. Appl. Microbiol. Biotechnol. 2016, 100, 9483–9497. [Google Scholar] [CrossRef] [PubMed]
  232. Tillmann, A.; Gow, N.A.; Brown, A.J. Nitric oxide and nitrosative stress tolerance in yeast. Biochem. Soc. Trans. 2011, 39, 219–223. [Google Scholar] [CrossRef]
  233. Shahani, N.; Sawa, A. Nitric oxide signaling and nitrosative stress in neurons: Role for S-nitrosylation. Antioxid. Redox Signal. 2011, 14, 1493–1504. [Google Scholar] [CrossRef] [PubMed]
  234. Ferrer-Sueta, G.; Campolo, N.; Trujillo, M.; Bartesaghi, S.; Carballal, S.; Romero, N.; Alvarez, B.; Radi, R. Biochemistry of Peroxynitrite and Protein Tyrosine Nitration. Chem. Rev. 2018, 118, 1338–1408. [Google Scholar] [CrossRef] [PubMed]
  235. Radi, R. Protein tyrosine nitration: Biochemical mechanisms and structural basis of functional effects. Acc. Chem. Res. 2013, 46, 550–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Wang, F.; Yuan, Q.; Chen, F.; Pang, J.; Pan, C.; Xu, F.; Chen, Y. Fundamental Mechanisms of the Cell Death Caused by Nitrosative Stress. Front. Cell Dev. Biol. 2021, 9, 742483. [Google Scholar] [CrossRef]
  237. Nakamura, T.; Tu, S.; Akhtar, M.W.; Sunico, C.R.; Okamoto, S.; Lipton, S.A. Aberrant protein s-nitrosylation in neurodegenerative diseases. Neuron 2013, 78, 596–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Seneviratne, U.; Nott, A.; Bhat, V.B.; Ravindra, K.C.; Wishnok, J.S.; Tsai, L.H.; Tannenbaum, S.R. S-nitrosation of proteins relevant to Alzheimer’s disease during early stages of neurodegeneration. Proc. Natl. Acad. Sci. USA 2016, 113, 4152–4157. [Google Scholar] [CrossRef] [Green Version]
  239. Han, F.; Chen, Y.X.; Lu, Y.M.; Huang, J.Y.; Zhang, G.S.; Tao, R.R.; Ji, Y.L.; Liao, M.H.; Fukunaga, K.; Qin, Z.H. Regulation of the ischemia-induced autophagy-lysosome processes by nitrosative stress in endothelial cells. J. Pineal Res. 2011, 51, 124–135. [Google Scholar] [CrossRef]
  240. Tripathi, D.N.; Chowdhury, R.; Trudel, L.J.; Tee, A.R.; Slack, R.S.; Walker, C.L.; Wogan, G.N. Reactive nitrogen species regulate autophagy through ATM-AMPK-TSC2-mediated suppression of mTORC1. Proc. Natl. Acad. Sci. USA 2013, 110, E2950–E2957. [Google Scholar] [CrossRef] [Green Version]
  241. Ma, L.L.; Ma, X.; Kong, F.J.; Guo, J.J.; Shi, H.T.; Zhu, J.B.; Zou, Y.Z.; Ge, J.B. Mammalian target of rapamycin inhibition attenuates myocardial ischaemia-reperfusion injury in hypertrophic heart. J. Cell. Mol. Med. 2018, 22, 1708–1719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Chakraborty, S.; Datta, S.; Ghosh, S. Induction of autophagy under nitrosative stress: A complex regulatory interplay between SIRT1 and AMPK in MCF7 cells. Cell. Signal. 2019, 64, 109411. [Google Scholar] [CrossRef] [PubMed]
  243. Porter, K.R.; Claude, A.; Fullam, E.F. A Study of Tissue Culture Cells by Electron Microscopy: Methods and Preliminary Observations. J. Exp. Med. 1945, 81, 233–246. [Google Scholar] [CrossRef] [PubMed]
  244. Wang, M.; Kaufman, R.J. Protein misfolding in the endoplasmic reticulum as a conduit to human disease. Nature 2016, 529, 326–335. [Google Scholar] [CrossRef]
  245. Hebert, D.N.; Molinari, M. In and out of the ER: Protein folding, quality control, degradation, and related human diseases. Physiol. Rev. 2007, 87, 1377–1408. [Google Scholar] [CrossRef]
  246. Merksamer, P.I.; Trusina, A.; Papa, F.R. Real-time redox measurements during endoplasmic reticulum stress reveal interlinked protein folding functions. Cell 2008, 135, 933–947. [Google Scholar] [CrossRef] [Green Version]
  247. Fujita, E.; Kouroku, Y.; Isoai, A.; Kumagai, H.; Misutani, A.; Matsuda, C.; Hayashi, Y.K.; Momoi, T. Two endoplasmic reticulum-associated degradation (ERAD) systems for the novel variant of the mutant dysferlin: Ubiquitin/proteasome ERAD(I) and autophagy/lysosome ERAD(II). Hum. Mol. Genet. 2007, 16, 618–629. [Google Scholar] [CrossRef]
  248. Schroder, M.; Kaufman, R.J. ER stress and the unfolded protein response. Mutat. Res. 2005, 569, 29–63. [Google Scholar] [CrossRef]
  249. Werner, E.D.; Brodsky, J.L.; McCracken, A.A. Proteasome-dependent endoplasmic reticulum-associated protein degradation: An unconventional route to a familiar fate. Proc. Natl. Acad. Sci. USA 1996, 93, 13797–13801. [Google Scholar] [CrossRef] [Green Version]
  250. Yorimitsu, T.; Nair, U.; Yang, Z.; Klionsky, D.J. Endoplasmic reticulum stress triggers autophagy. J. Biol. Chem. 2006, 281, 30299–30304. [Google Scholar] [CrossRef] [Green Version]
  251. Bernales, S.; McDonald, K.L.; Walter, P. Autophagy counterbalances endoplasmic reticulum expansion during the unfolded protein response. PLoS Biol. 2006, 4, e423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Welihinda, A.A.; Kaufman, R.J. The unfolded protein response pathway in Saccharomyces cerevisiae: Oligomerization and trans-phosphorylation of Ire1p (Ern1p) are required for kinase activation. J. Biol. Chem. 1996, 271, 18181–18187. [Google Scholar] [PubMed] [Green Version]
  253. Okamura, K.; Kimata, Y.; Higashio, H.; Tsuru, A.; Kohno, K. Dissociation of Kar2p/BiP from an ER sensory molecule, Ire1p, triggers the unfolded protein response in yeast. Biochem. Biophys. Res. Commun. 2000, 279, 445–450. [Google Scholar] [CrossRef] [PubMed]
  254. Cox, J.S.; Walter, P. A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell 1996, 87, 391–404. [Google Scholar] [CrossRef] [Green Version]
  255. Sidrauski, C.; Walter, P. The transmembrane kinase Ire1p is a site-specific endonuclease that initiates mRNA splicing in the unfolded protein response. Cell 1997, 90, 1031–1039. [Google Scholar] [PubMed] [Green Version]
  256. Sidrauski, C.; Cox, J.S.; Walter, P. tRNA ligase is required for regulated mRNA splicing in the unfolded protein response. Cell 1996, 87, 405–413. [Google Scholar]
  257. Mori, K. Signalling pathways in the unfolded protein response: Development from yeast to mammals. J. Biochem. 2009, 146, 743–750. [Google Scholar] [CrossRef]
  258. Kawahara, T.; Yanagi, H.; Yura, T.; Mori, K. Endoplasmic reticulum stress-induced mRNA splicing permits synthesis of transcription factor Hac1p/Ern4p that activates the unfolded protein response. Mol. Biol. Cell 1997, 8, 1845–1862. [Google Scholar] [CrossRef] [Green Version]
  259. Patil, C.; Walter, P. Intracellular signaling from the endoplasmic reticulum to the nucleus: The unfolded protein response in yeast and mammals. Curr. Opin. Cell Biol. 2001, 13, 349–355. [Google Scholar] [CrossRef]
  260. Chakraborty, R.; Baek, J.H.; Bae, E.Y.; Kim, W.-Y.; Lee, S.Y.; Kim, M.G. Comparison and contrast of plant, yeast, and mammalian ER stress and UPR. Appl. Biol. Chem. 2016, 59, 337–347. [Google Scholar] [CrossRef]
  261. Reddy, R.K.; Mao, C.; Baumeister, P.; Austin, R.C.; Kaufman, R.J.; Lee, A.S. Endoplasmic reticulum chaperone protein GRP78 protects cells from apoptosis induced by topoisomerase inhibitors: Role of ATP binding site in suppression of caspase-7 activation. J. Biol. Chem. 2003, 278, 20915–20924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Gardner, B.M.; Walter, P. Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response. Science 2011, 333, 1891–1894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Calfon, M.; Zeng, H.; Urano, F.; Till, J.H.; Hubbard, S.R.; Harding, H.P.; Clark, S.G.; Ron, D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 2002, 415, 92–96. [Google Scholar] [CrossRef] [PubMed]
  264. Acosta-Alvear, D.; Zhou, Y.; Blais, A.; Tsikitis, M.; Lents, N.H.; Arias, C.; Lennon, C.J.; Kluger, Y.; Dynlacht, B.D. XBP1 controls diverse cell type-and condition-specific transcriptional regulatory networks. Mol. Cell 2007, 27, 53–66. [Google Scholar] [CrossRef] [PubMed]
  265. Margariti, A.; Li, H.; Chen, T.; Martin, D.; Vizcay-Barrena, G.; Alam, S.; Karamariti, E.; Xiao, Q.; Zampetaki, A.; Zhang, Z. XBP1 mRNA splicing triggers an autophagic response in endothelial cells through BECLIN-1 transcriptional activation. J. Biol. Chem. 2013, 288, 859–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Zhou, Y.; Lee, J.; Reno, C.M.; Sun, C.; Park, S.W.; Chung, J.; Lee, J.; Fisher, S.J.; White, M.F.; Biddinger, S.B. Regulation of glucose homeostasis through a XBP-1–FoxO1 interaction. Nat. Med. 2011, 17, 356–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Vidal, R.L.; Figueroa, A.; Court, F.A.; Thielen, P.; Molina, C.; Wirth, C.; Caballero, B.; Kiffin, R.; Segura-Aguilar, J.; Cuervo, A.M. Targeting the UPR transcription factor XBP1 protects against Huntington’s disease through the regulation of FoxO1 and autophagy. Hum. Mol. Genet. 2012, 21, 2245–2262. [Google Scholar] [CrossRef]
  268. Zhao, Y.; Li, X.; Cai, M.-Y.; Ma, K.; Yang, J.; Zhou, J.; Fu, W.; Wei, F.-Z.; Wang, L.; Xie, D. XBP-1u suppresses autophagy by promoting the degradation of FoxO1 in cancer cells. Cell Res. 2013, 23, 491–507. [Google Scholar] [CrossRef] [Green Version]
  269. Li, D.D.; Wang, L.L.; Deng, R.; Tang, J.; Shen, Y.; Guo, J.F.; Wang, Y.; Xia, L.P.; Feng, G.K.; Liu, Q.Q.; et al. The pivotal role of c-Jun NH2-terminal kinase-mediated Beclin 1 expression during anticancer agents-induced autophagy in cancer cells. Oncogene 2009, 28, 886–898. [Google Scholar] [CrossRef] [Green Version]
  270. Meares, G.P.; Hughes, K.J.; Naatz, A.; Papa, F.R.; Urano, F.; Hansen, P.A.; Benveniste, E.N.; Corbett, J.A. IRE1-dependent activation of AMPK in response to nitric oxide. Mol. Cell. Biol. 2011, 31, 4286–4297. [Google Scholar] [CrossRef] [Green Version]
  271. Kouroku, Y.; Fujita, E.; Tanida, I.; Ueno, T.; Isoai, A.; Kumagai, H.; Ogawa, S.; Kaufman, R.J.; Kominami, E.; Momoi, T. ER stress (PERK/eIF2α phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007, 14, 230–239. [Google Scholar] [CrossRef] [Green Version]
  272. Pakos-Zebrucka, K.; Koryga, I.; Mnich, K.; Ljujic, M.; Samali, A.; Gorman, A.M. The integrated stress response. EMBO Rep. 2016, 17, 1374–1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Pattingre, S.; Tassa, A.; Qu, X.; Garuti, R.; Liang, X.H.; Mizushima, N.; Packer, M.; Schneider, M.D.; Levine, B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005, 122, 927–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Rzymski, T.; Milani, M.; Pike, L.; Buffa, F.; Mellor, H.; Winchester, L.; Pires, I.; Hammond, E.; Ragoussis, I.; Harris, A. Regulation of autophagy by ATF4 in response to severe hypoxia. Oncogene 2010, 29, 4424–4435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Avivar-Valderas, A.; Bobrovnikova-Marjon, E.; Diehl, J.A.; Bardeesy, N.; Debnath, J.; Aguirre-Ghiso, J. Regulation of autophagy during ECM detachment is linked to a selective inhibition of mTORC1 by PERK. Oncogene 2013, 32, 4932–4940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Brüning, A.; Rahmeh, M.; Friese, K. Nelfinavir and bortezomib inhibit mTOR activity via ATF4-mediated sestrin-2 regulation. Mol. Oncol. 2013, 7, 1012–1018. [Google Scholar] [CrossRef] [PubMed]
  277. Adachi, Y.; Yamamoto, K.; Okada, T.; Yoshida, H.; Harada, A.; Mori, K. ATF6 is a transcription factor specializing in the regulation of quality control proteins in the endoplasmic reticulum. Cell Struct. Funct. 2008, 33, 75–89. [Google Scholar] [CrossRef] [Green Version]
  278. Guo, F.-J.; Xiong, Z.; Lu, X.; Ye, M.; Han, X.; Jiang, R. ATF6 upregulates XBP1S and inhibits ER stress-mediated apoptosis in osteoarthritis cartilage. Cell. Signal. 2014, 26, 332–342. [Google Scholar] [CrossRef]
  279. Hirsch, I.; Weiwad, M.; Prell, E.; Ferrari, D.M. ERp29 deficiency affects sensitivity to apoptosis via impairment of the ATF6–CHOP pathway of stress response. Apoptosis 2014, 19, 801–815. [Google Scholar] [CrossRef]
  280. Yung, H.W.; Charnock-Jones, D.S.; Burton, G.J. Regulation of AKT phosphorylation at Ser473 and Thr308 by endoplasmic reticulum stress modulates substrate specificity in a severity dependent manner. PLoS ONE 2011, 6, e17894. [Google Scholar] [CrossRef]
  281. Kalvakolanu, D.V.; Gade, P. IFNG and autophagy: A critical role for the ER-stress mediator ATF6 in controlling bacterial infections. Autophagy 2012, 8, 1673–1674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Gade, P.; Manjegowda, S.B.; Nallar, S.C.; Maachani, U.B.; Cross, A.S.; Kalvakolanu, D.V. Regulation of the death-associated protein kinase 1 expression and autophagy via ATF6 requires apoptosis signal-regulating kinase 1. Mol. Cell. Biol. 2014, 34, 4033–4048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Høyer-Hansen, M.; Bastholm, L.; Szyniarowski, P.; Campanella, M.; Szabadkai, G.; Farkas, T.; Bianchi, K.; Fehrenbacher, N.; Elling, F.; Rizzuto, R. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-β, and Bcl-2. Mol. Cell 2007, 25, 193–205. [Google Scholar] [CrossRef] [PubMed]
  284. Borodkina, A.V.; Shatrova, A.N.; Deryabin, P.I.; Griukova, A.A.; Abushik, P.A.; Antonov, S.M.; Nikolsky, N.N.; Burova, E.B. Calcium alterations signal either to senescence or to autophagy induction in stem cells upon oxidative stress. Aging 2016, 8, 3400–3418. [Google Scholar] [CrossRef] [Green Version]
  285. Simon, B.; Huart, A.-S.; Temmerman, K.; Vahokoski, J.; Mertens, H.D.; Komadina, D.; Hoffmann, J.-E.; Yumerefendi, H.; Svergun, D.I.; Kursula, P. Death-associated protein kinase activity is regulated by coupled calcium/calmodulin binding to two distinct sites. Structure 2016, 24, 851–861. [Google Scholar] [CrossRef] [Green Version]
  286. Gomes, L.R.; Menck, C.F.M.; Leandro, G.S. Autophagy Roles in the Modulation of DNA Repair Pathways. Int. J. Mol. Sci. 2017, 18, 2351. [Google Scholar] [CrossRef] [Green Version]
  287. Sharma, V.; Verma, S.; Seranova, E.; Sarkar, S.; Kumar, D. Selective Autophagy and Xenophagy in Infection and Disease. Front. Cell Dev. Biol. 2018, 6, 147. [Google Scholar] [CrossRef] [Green Version]
  288. Feng, Y.; Yao, Z.; Klionsky, D.J. How to control self-digestion: Transcriptional, post-transcriptional, and post-translational regulation of autophagy. Trends Cell Biol. 2015, 25, 354–363. [Google Scholar] [CrossRef] [Green Version]
  289. Holczer, M.; Hajdu, B.; Lorincz, T.; Szarka, A.; Banhegyi, G.; Kapuy, O. Fine-tuning of AMPK-ULK1-mTORC1 regulatory triangle is crucial for autophagy oscillation. Sci. Rep. 2020, 10, 17803. [Google Scholar] [CrossRef]
  290. Ling, N.X.Y.; Kaczmarek, A.; Hoque, A.; Davie, E.; Ngoei, K.R.W.; Morrison, K.R.; Smiles, W.J.; Forte, G.M.; Wang, T.; Lie, S.; et al. mTORC1 directly inhibits AMPK to promote cell proliferation under nutrient stress. Nat. Metab. 2020, 2, 41–49. [Google Scholar] [CrossRef]
  291. He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef] [PubMed]
  292. Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef] [PubMed]
  293. Ghemrawi, R.; Khair, M. Endoplasmic Reticulum Stress and Unfolded Protein Response in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 6127. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Autophagy in yeast and mammalian systems. (A) Four stages of autophagy. The upper and lower parts of each panel represent yeast and mammals, respectively. In both yeast and mammals, autophagy includes four stages, induction and nucleation of the phagophore, expansion and maturation of the phagophore, fusion with the vacuole (in yeast)/lysosome (in mammals), and degradation and efflux of the breakdown products. (B) Protein complexes involved in induction and nucleation of the phagophore. In yeast, Atg1 and PtdIns3K complex I will be recruited to the PAS and drive the formation of PtdIns3P on the phagophore. In the mammalian system, the ULK1 complex phosphorylates and activates PtdIns3K complex I, which contributes to the formation of the phagophore. (C) Two ubiquitin-like systems. In both yeast and mammals, the Atg12 complex (Atg12–Atg5-Atg16 in yeast and ATG12–ATG5-ATG16L1 in mammals) forms with the help of Atg7/ATG7 and Atg10/ATG10; this complex then functions as an E3 enzyme for the conjugation of Atg8 (in yeast) and Atg8-family proteins (LC3 and GABARAP subfamilies in mammals) with PE.
Figure 1. Autophagy in yeast and mammalian systems. (A) Four stages of autophagy. The upper and lower parts of each panel represent yeast and mammals, respectively. In both yeast and mammals, autophagy includes four stages, induction and nucleation of the phagophore, expansion and maturation of the phagophore, fusion with the vacuole (in yeast)/lysosome (in mammals), and degradation and efflux of the breakdown products. (B) Protein complexes involved in induction and nucleation of the phagophore. In yeast, Atg1 and PtdIns3K complex I will be recruited to the PAS and drive the formation of PtdIns3P on the phagophore. In the mammalian system, the ULK1 complex phosphorylates and activates PtdIns3K complex I, which contributes to the formation of the phagophore. (C) Two ubiquitin-like systems. In both yeast and mammals, the Atg12 complex (Atg12–Atg5-Atg16 in yeast and ATG12–ATG5-ATG16L1 in mammals) forms with the help of Atg7/ATG7 and Atg10/ATG10; this complex then functions as an E3 enzyme for the conjugation of Atg8 (in yeast) and Atg8-family proteins (LC3 and GABARAP subfamilies in mammals) with PE.
Antioxidants 11 00304 g001
Figure 2. Autophagy regulation by TORC1 and MTORC1. (A) In yeast, nutrient sources activate TORC1 basically through activated Gtr1-Gtr2 heterodimer. The activated MTORC1 inhibits auto-phagy through phosphorylating Atg13, inducing degradation of ATG mRNA and inhibiting Rim15 translocation to nucleus, which releases some repressing transcription factors such as Rph1 and Ume6 and induces ATG genes transcription. (B) In mammalian cells, amino acids activate RRAG GTPase and recruit MTORC1 to lysosomes, where it is activated by RHEB. Activated MTORC1 phosphorylates several ATG proteins and inhibits their functions. Meanwhile, MTORC1 dependent phosphorylation in TFEB, MITF, and TFE3 blocks their translocation into nucleus and the induction of ATG and CLEAR genes transcription. Solid and dashed lines represent direct and indirect regulations respectively.
Figure 2. Autophagy regulation by TORC1 and MTORC1. (A) In yeast, nutrient sources activate TORC1 basically through activated Gtr1-Gtr2 heterodimer. The activated MTORC1 inhibits auto-phagy through phosphorylating Atg13, inducing degradation of ATG mRNA and inhibiting Rim15 translocation to nucleus, which releases some repressing transcription factors such as Rph1 and Ume6 and induces ATG genes transcription. (B) In mammalian cells, amino acids activate RRAG GTPase and recruit MTORC1 to lysosomes, where it is activated by RHEB. Activated MTORC1 phosphorylates several ATG proteins and inhibits their functions. Meanwhile, MTORC1 dependent phosphorylation in TFEB, MITF, and TFE3 blocks their translocation into nucleus and the induction of ATG and CLEAR genes transcription. Solid and dashed lines represent direct and indirect regulations respectively.
Antioxidants 11 00304 g002
Figure 3. AMPK drives autophagy through three layers of regulation. (1) AMPK suppresses MTOR activity by phosphorylating TSC2 and RPTOR. (2) AMPK directly phosphorylates and activates proteins involved in autophagy including ULK1, BECN1, PIK3C3/VPS34, and ATG9A. (3) AMPK activates the positive regulators of autophagy, for example, phosphorylation of FOXO3 leads to increased transcription of autophagy-related genes.
Figure 3. AMPK drives autophagy through three layers of regulation. (1) AMPK suppresses MTOR activity by phosphorylating TSC2 and RPTOR. (2) AMPK directly phosphorylates and activates proteins involved in autophagy including ULK1, BECN1, PIK3C3/VPS34, and ATG9A. (3) AMPK activates the positive regulators of autophagy, for example, phosphorylation of FOXO3 leads to increased transcription of autophagy-related genes.
Antioxidants 11 00304 g003
Figure 4. The transcriptional regulation of autophagy by oxidative stress. (A) In yeast, upon oxidative stress, oxidized Yap1 is accumulated in the nucleus and drives the expression of detoxifying enzymes and the autophagy-related gene ATG15. YRE: Yap response element. (B) In mammalian cells, upon oxidative stress, the oxidation of cysteine residues of KEAP1 prevent the ubiquitination of NFE2L2/NRF2, thus allowing NFE2L2 to enter the nucleus and activate several autophagy genes. ARE: antioxidant response element. (C) In pathological conditions, nitrosative stress activates autophagy activity via multiple signaling cascades.
Figure 4. The transcriptional regulation of autophagy by oxidative stress. (A) In yeast, upon oxidative stress, oxidized Yap1 is accumulated in the nucleus and drives the expression of detoxifying enzymes and the autophagy-related gene ATG15. YRE: Yap response element. (B) In mammalian cells, upon oxidative stress, the oxidation of cysteine residues of KEAP1 prevent the ubiquitination of NFE2L2/NRF2, thus allowing NFE2L2 to enter the nucleus and activate several autophagy genes. ARE: antioxidant response element. (C) In pathological conditions, nitrosative stress activates autophagy activity via multiple signaling cascades.
Antioxidants 11 00304 g004
Figure 5. Summary of autophagy regulation under stress. Mammalian systems are presented here to demonstrate the major causes of stress, the main stress-sensing molecules and pathways and their regulation of autophagy.
Figure 5. Summary of autophagy regulation under stress. Mammalian systems are presented here to demonstrate the major causes of stress, the main stress-sensing molecules and pathways and their regulation of autophagy.
Antioxidants 11 00304 g005
Table 1. Stress biomarkers and their detection.
Table 1. Stress biomarkers and their detection.
Stress TypeOrganismBiomarkersDetectionReference
Nutrient stressYeastTORC1
inactivation
Sch9 dephosphorylation[27]
Mammalian EIF4EBP1 dephosphorylation in vitro[28]
MammalsMTORC1
inactivation
RPS6KB1 dephosphorylation[29]
EIF4EBP1 dephosphorylation[29]
Energy stressYeast and mammalsLower ATP: ADP/AMP ratioLiquid chromatography to detect ATP, ADP and AMP level[30]
ATP:ADP fluorescence reporter[31,32]
Bioluminescent detection[33]
YeastSnf1 activation“SAMS” peptide phosphorylation[34]
MammalsAMPK activationAMPK phosphorylation[35]
Phosphorylation of downstream targets such as ACAC (acetyl-CoA carboxylase)[35]
Oxidative stressYeast and MammalsHigh level of ROSDichlorodihydrofluorescein fluorescence[36]
PG1 or PC1 fluorescence[37]
Calcein-acetoxymethylester (calcein-AM) fluorescence[38]
CellROX dye[39]
Increased GSSG:GSH ratioHigh-performance liquid chromatography[40]
Capillary electrophoresis[40]
Bioluminescence[41]
Genetically-encoded fluorescent sensors[42,43]
Lipid
peroxidation
Fluorescence shift of C11-BODIPY (581/591)[44]
TBA-MDA assay[45,46]
ER stressYeastMisfolded protein accumulationKar2 sedimentation[47]
UPR pathway
activation
Transcription reporter containing a UPR element promoter driving fluorescent proteins [47]
Ire1 clustering[47]
MammalsProtein
aggregates
Thioflavin T (ThT)
fluorescence
[48]
UPR pathway
activation
Spliced XBP1 mRNA detection using ER stress-activated indicator” (ERAI) construct[49]
Upregulated expression of UPR target genes, including DDIT3 and HSPA5/GRP78[50]
ATF6 translocation[51]
Table 2. Autophagy regulation in yeast under nutrient-rich and starvation conditions.
Table 2. Autophagy regulation in yeast under nutrient-rich and starvation conditions.
Type of RegulationRegulatory
Factors
ConditionsEffects (↑, Positive; ↓, Negative)Target Genes or
Proteins
Reference
Transcriptional regulationPho23Nutrient-richATG1, 7, 8, 9, 12, 14, 29[69]
Spt10Nutrient-richATG1, 7, 9,14, 32[62]
Fyv5Nutrient-rich and starvationATG1, 7, 8, 9,14, 29, 32
Sfl1Nutrient-richATG1, 7, 8, 9, 14, 29, 32
Sko1Nutrient-rich and starvationATG1, 7, 8, 32
Zap1Nutrient-richATG1, 7, 8, 9, 14, 29, 32
Swi5Nutrient-richATG7, 8, 9, 14, 29
Rsc1StarvationATG8[70]
Spt4/5Nutrient-richATG8, 41[71]
StarvationATG41
Post-transcriptional regulationXrn1Nutrient-richATG1, 4, 5, 7, 8, 12, 14, 16, 29, 31[72]
Dhh1Nutrient-richATG3, 7, 8, 19, 20, 22, 24[68]
StarvationATG1,13[73]
Pat1StarvationATG1, 2,7, 9[74]
Psp2StarvationATG1, 13[75]
Ded1StarvationATG1[76]
Post-
translational regulation
PhosphorylationHrr25Cvt pathway and
pexophagy
induction
Atg19,36[77]
UbiquitinationMet30Nutrient-richAtg9[78]
AcetylationEsa1StarvationAtg3[79]
DeacetylationRpd3StarvationAtg3[79]
Epigenetic
regulation
AcetylationSas2Nutrient-richHistone H4 Lys16[80]
MethylationUnclearNutrient-richHistone H3 Lys4[80]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lei, Y.; Huang, Y.; Wen, X.; Yin, Z.; Zhang, Z.; Klionsky, D.J. How Cells Deal with the Fluctuating Environment: Autophagy Regulation under Stress in Yeast and Mammalian Systems. Antioxidants 2022, 11, 304. https://doi.org/10.3390/antiox11020304

AMA Style

Lei Y, Huang Y, Wen X, Yin Z, Zhang Z, Klionsky DJ. How Cells Deal with the Fluctuating Environment: Autophagy Regulation under Stress in Yeast and Mammalian Systems. Antioxidants. 2022; 11(2):304. https://doi.org/10.3390/antiox11020304

Chicago/Turabian Style

Lei, Yuchen, Yuxiang Huang, Xin Wen, Zhangyuan Yin, Zhihai Zhang, and Daniel J. Klionsky. 2022. "How Cells Deal with the Fluctuating Environment: Autophagy Regulation under Stress in Yeast and Mammalian Systems" Antioxidants 11, no. 2: 304. https://doi.org/10.3390/antiox11020304

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

Article Metrics

Back to TopTop