Autophagy
Mary Johnson (han at labome dot com)
Synatom Research, Princeton, New Jersey, United States
DOI
//dx.doi.org/10.13070/mm.en.8.2672
Date
last modified : 2023-05-21; original version : 2018-11-26
Cite as
MATER METHODS 2018;8:2672

Autophagy, meaning “self-eating” in Greek, is an evolutionarily conserved cellular degradative mechanism in eukaryotes. Autophagy maintains cellular homeostasis by degrading unwanted cellular materials in the lysosomes; and thus eliminates harmful contents such as intracellular pathogens including bacteria, viruses, fungi, and protozoa, damaged or surplus organelles, and harmful protein aggregates. The role of autophagy is not limited to the elimination of cargo - it plays a regulatory role in other processes such as innate and adaptive immunity, inflammation, development, diseases such as neurodegenerative diseases, cancer, metabolic diseases, and cardiovascular diseases among others [1]. For his pioneering work on autophagy, Dr. Ohsumi Yoshinori won the Nobel Prize in 2016 which underscores the importance of autophagy in health and diseases. Various pre-clinical and clinical studies have explored whether manipulation of autophagy can be used for treating various diseases in animal models.

Protein Top three suppliers Reference
ATG5 autophagy-related 5Cell Signaling Technology 12994 (49), Abcam ab108327 (21), Santa Cruz Biotechnology sc-133158 (8) [2]
ATG7 autophagy-related 7Cell Signaling Technology 8558 (58), Abcam ab52472 (7), Santa Cruz Biotechnology sc-376212 (6) [2]
ATG9A autophagy-related 9AAbcam ab108338 (8), Cell Signaling Technology 13509 (6), Invitrogen MA1-149 (1) [2]
ATG14 / ATG14L autophagy-related 14MBL International PD026 (14) [3]
ATG16L1 autophagy related 16 like 1Cell Signaling Technology 8089 (24), MBL International M150-3 (6), Abcam ab187671 (4) [2]
CALCOCO2 / NDP52 calcium binding and coiled-coil domain 2Santa Cruz Biotechnology sc-376540 (3), OriGene CF501971 (1) [2]
LAMP2 lysosomal associated membrane protein 2Santa Cruz Biotechnology sc-18822 (46), Developmental Studies Hybridoma Bank H4B4 (35), Abcam ab25631 (30) [4]
MAP1LC3B / LC3 microtubule associated protein 1 light chain 3 betaCell Signaling Technology 3868 (171), Abcam ab192890 (14), MilliporeSigma SAB4200361 (6) [2, 5]
OPTN optineurinSanta Cruz Biotechnology sc-166576 (9), Cell Signaling Technology 58981 (2) [2]
PIK3C3 / VPS34 phosphatidylinositol 3-kinase catalytic subunit type 3Cell Signaling Technology 4263 (31), Echelon Biosciences Z-R015 (7), Abcam ab124905 (3) [3]
PIK3R4 / VPS15Abnova H00030849-M02 (3), Abcam ab128903 (1) [2, 3]
PINK1 PTEN induced kinase 1Cell Signaling Technology 6946 (21), OriGene TA324354 (1), Santa Cruz Biotechnology sc-518052 (1) [2]
PRKN parkin RBR E3 ubiquitin protein ligaseSanta Cruz Biotechnology sc-32282 (48), Cell Signaling Technology 4211 (29), Abcam ab77924 (16) [2]
RETREG1 / FAM134B reticulophagy regulator 1Abcam ab151755 (2) [2]
RUBCN RUN and cysteine rich domain containing beclin 1 interacting proteinCell Signaling Technology 8465 (8), MBL International M170-3 (1) [2]
SQSTM1 sequestosome 1Abcam ab56416 (99), Santa Cruz Biotechnology sc-28359 (73), Abnova H00008878-M01 (46) [2]
TBK1 TANK binding kinase 1Cell Signaling Technology 5483 (96), Abcam ab40676 (22), Novus Biologicals NB100-56705 (6) [2]
ULK1 unc-51 like autophagy activating kinase 1Cell Signaling Technology 8054 (79), Santa Cruz Biotechnology sc-390904 (3), Abcam ab133747 (1) [2]
WIPI2 WD repeat domain, phosphoinositide interacting 2Abcam ab105459 (10), Bio-Rad MCA5780GA (8) [2, 3]
WIPI3 WD repeat domain 45BSanta Cruz Biotechnology sc-514194 (2) [2]
Table 1. Human autophagy-related proteins and top cited antibodies against them among the over 60,000 formal publications in Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.

Three types of autophagy exist: macroautophagy, microautophagy and chaperone-mediated autophagy. Macroautophagy encapsulates the cargo for degradation in a double-membrane structure called autophagosome which fuses with the lysosome and delivers the contents for degradation. Microautophagy mediates direct capturing of the cargo by lysosomes without the formation of autophagosomes [6]. Chaperone-mediated autophagy involves selective degradation of the target proteins directly into the lysosome which is delivered by a chaperone [7]. Of these, the most well-studied macroautophagy (hereafter called autophagy) includes mitophagy (for mitochondria), ERphagy (for endoplasmic reticulum), pexophagy (for peroxisomes), nucleophagy (for nucleus), ribophagy (for ribosomes), aggrephagy (for aggregates) and xenophagy (for pathogens). Here, we discuss the process of autophagy and some of the common methods for monitoring autophagy in vitro and in vivo.

Process of Autophagy

Autophagy is initiated with the formation of a double-membrane structure called autophagosome around the cellular material earmarked for degradation. Autophagosome then delivers its contents to the lysosome by fusing with the lysosomes to generate autolysosomes where the contents are degraded by the activity of proteolytic enzymes. Multiple proteins are involved in authophagy (Table 1) and new involved proteins are being identified - the COPII-cargo adaptor complex Lst1-Sec23 was found to be involved in the ER-phagy [8]. Autophagy is initiated by ULK1, Beclin1 and PI3K-III complexes that promote the formation of an isolation membrane [9]. The isolation membrane gets decorated with LC3 (a mammalian homolog of yeast Atg8) which serves as an anchor for recruiting the selected cargo. LC3 is first cleaved by ATG4 to yield LC3-I revealing the glycine residue on LC3. Two ubiquitin-like conjugation systems ATG7-ATG3 and ATG5-ATG12-ATG16 further facilitate the recruitment of LC3-I to the autophagosomal membrane by conjugating LC3-I to lipid PE molecules to generate LC3-II which is recruited to the inner and the outer membranes of the autophagosomes [10]. Proteins such as p62 act as adaptor receptors that dock the cargo to the autophagosome by binding to the cargo on one end and to LC3-II on the other and deliver the cargo to the autophagosomes [11]. Fusion of autophagosome with the lysosome yields autolysosomes and this process is mediated by a Soluble NSF Attachment Protein Receptor (SNARE) protein syntaxin 17 [12]. The inner membrane of the autolysosome is subsequently degraded which includes LC3-II bound to the inner membrane while LC3-II on the outer membrane is released and recycled by the activity of ATG4. The process of cargo selection and degradation is referred to as autophagy flux and is indicative of functional and complete autophagy. Some of the well-studied inducers and inhibitors of autophagy are listed in Table 2, 3, respectively.

Inducer Function Reference
RapamycinInhibits mTOR [4, 13]
TamoxifenIncrease lysosomal pH thereby preventing autolysosomal degradation
ResveratrolmTOR inhibitor
EverolimusmTOR inhibitor
TrehalosemTOR-independent autophagy activator
CurcuminmTOR inhibitor, increases Atg8
RottlerinmTOR inhibitor
Vitamin D3Increases LC3 and Beclin1
SpermidineAcetylase inhibitor
Table 2. Autophagy inducers. New ones such as OSI-027, AZD2014, AZD8055 and others have also been identified [4].
Methods to Detect Autophagy

Several "standard" methods of monitoring autophagy are discussed below and summarized in Table 4. Often several methods are combined together to detect autophagy. For example, Saito T et al used the conversion of LC3-I to LC3-II and p62 degradation in Western blots as indicators of autophagy [14].

Inhibitor Function Reference
3-MethyladenineA class III  PtdIns3K inhibitor that blocks  early stage of autophagy. Not a specific autophagy inhibitor [4, 5]
Bafilomycin A1A V-ATPase inhibitor that blocks fusion of autophagosomes with the lysosomes [4, 15]
Chloroquine, NH4ClIncrease lysosomal pH thereby preventing autolysosomal degradation [4, 13]
Colchicineblock autophagosome maturation to autolysosomes [16]
E64dInhibitor of lysosomal proteases. Usually used along with pepstatin A [17]
LeupeptinInhibitor of lysosomal proteases. Usually used along with pepstatin A and E64d
Pepstatin AInhibitor of lysosomal proteases. Usually used along with E64d
siRNA, miRNA, shRNA against Atg genesBlock expression of autophagy gene
WortmanninInhibits PtdIns3K to block autophagy. Not a specific inhibitor
LY294002PI3 Kinase inhibitor
VinblastineInhibits autophagosome-lysosome fusion
Table 3. Autophagy inhibitors.
LC3 II turnover

Multiple approaches can be used to study the LC3 II turnover.

Western blotting

Upon induction of autophagy, LC3-I is lipidated to become LC3-II which migrates faster than its parent molecule on an SDS-PAGE gel. LC3I and LC3II migrate as ~16 and ~14Kda proteins respectively. Thus, increase in LC3-II level has been considered as a gold standard for demonstrating autophagic induction. However, as LC3-II is subsequently degraded by the activity of proteases in the autolysosomes (a process referred to as autophagy flux), the increase in LC3-II by a test compound may not necessarily reflect autophagy activation but may also suggest a block in autophagy flux [18, 19]. To distinguish between these two possible outcomes, one uses inhibitors of lysosome-dependent degradation such as Bafilomycin A1, E64d, pepstatin A, and leupeptin that either prevent the fusion of autophagosomes with autolysosome or block the proteolytic activity of the degradative enzymes in the autolysosomes. For example, Ling Q et al treated Arabidopsis protoplasts with 1-10 uM E64 from Melford for 2 hours to inhibit autophagy [17]. Using these inhibitors, the following outcomes may be possible when testing a reagent for its ability to induce autophagy.

  • The treatment causes an increased amount of LC3-II in the presence or absence of the inhibitors when compared to the untreated controls. This would suggest that autophagy flux is increased in this scenario.
  • The treatment causes no change in LC3-II expression with or without the flux inhibitors in comparison to the untreated samples. This suggests that autophagy is inhibited under these conditions.
  • The treatment causes increased LC3-II in the absence but not in the presence of the inhibitors. This suggests that under this condition, autophagy flux is suppressed.

Finally, as LC3 antibodies have an overall greater tendency for LC3-II compared to LC3-I, it is not recommended quantifying the LC3-II/LC3-I ratio, although it is still commonly used [20]. Instead, one should measure the LC3-II/actin or other housekeeping gene ratio when quantifying immunoblots [21]. Autophagy induction must also be validated by using genetic inhibitors such as siRNAs against autophagy genes such as Atg5, Atg7, or Beclin1 and/or by using pharmacological inhibitors such as 3-methyladenine (3 MA), or conditional knockout of genes like ATG7 [22]. Some of these inhibitors are discussed in Table 3. While testing the ability of a test reagent to induce autophagy, one should also use known autophagy inducers and inhibitors as positive and negative controls respectively in their studies. For instance, Tamaki Y et al used bafilomycin at 0.1 μM in HEK293A cell culture to demonstrate the involvement of autophagy–lysosome activity in intrabody-induced TDP-43 protein degradation [23].

Method Description Reference
LC3 western blottingLC3II is detected using anti-LC3 antibodies in the presence or absence of a lysosomal inhibitor such as bafilomycin A1 by SDS-PAGE. [4, 15]
LC3 fluorescence microscopyIncrease in the LC3 puncta is detected by using RFP-GFP-LC3 or GFP-LC3 expressing plasmid, or a tandem mRFP-eGFP-LC3 transgene, or by anti-LC3 antibodies for detecting endogenous LC3 protein. Similar techniques include galectin puncta formation assay and fluorescent dextran release assay [24]. [3, 25]
P62 degradationP62 degradation is monitored in the absence or presence of lysosomal inhibitor by western blotting using anti-p62 antibody. [4, 14]
Flow cytometryLoss of GFP signal in GFP-LC3 expressing  cells, increase in the accumulation of autophagosome-specific dyes [26, 27]
Image cytometryIn combination with acridine orange staining to study late-stage autophagy [28]
Turnover of long-lived proteinsDone using radioactive or non-radioactive amino acids in the absence and presence of a lysosomal inhibitor [27, 29]
Electron microscopyUsed for monitoring the autophagic structures, the autophagosome number and the contents of autophagosomes. New fixation techniques improve visualization of autophagosomes [30]. [31]
Autophagy gene and protein expressionQuantitative PCR, Northern blot, Western blotting [32]
KeimaDetection of autophagosomes using a fluorescence protein with a bimodal excitation spectrum, by fluorescence microscopy [33]
Cargo Sequestration assayMeasurement of bulk autophagy using a cargo marker, which represents total cytoplasm. [34]
Semiconductor-based in situ measurementA semiconductor-based field-effect transistor (FET) biosensor senses changes of ionic or molecular charges. [35]
Table 4. Summary of methods used for monitoring autophagy.
Fluorescence microscopy

LC3-II can also be monitored by fluorescence microscopy and can be distinguished from the parent LC3-I molecule due to its ability to become membrane bound via its lipidated moiety. Thus, while LC3-I is cytosolic, membrane-bound LC3-II represents autophagosomes that are visible as dots or “puncta”. Detection of puncta can be accomplished either by using antibodies against endogenous LC3 or by using GFP-LC3 expressing plasmid [27, 36]. While an increase in the number of GFP-LC3 puncta was considered to be indicative of an increase in autophagy in the past, now, the same criteria of using inhibitors such as bafilomycin A1 should be used to distinguish between the activation of autophagy and the suppression of autophagy flux, when using LC3 puncta to quantitate the autophagosomes.

A more advanced method for measuring autophagy flux by LC3 puncta is to use tandem labeled fluorescent LC3 expression vectors such as mRFP-GFP-LC3 [27] or mCherry-EGFP-LC3 plasmids [37]. EGFP-LC3 plasmid was initially developed as one of earlier probes to monitor autophagosome formation in the cell [36]. However, it presents significant limitations as GFP is low pH-sensitive and is quenched in the autolysosomes. Thus, information pertaining to the autophagic flux is not possible with the use of GFP-LC3 unless inhibitors such as bafilomycin A1 are used. RFP or cherry molecule (red) on the other hand is more stable in low pH autolysosomes and thus red puncta would reflect only autolysosomes while the yellow puncta (GFP and RFP signal) would reflect autophagosomes thereby providing a detailed analysis of autophagy flux. For example, Vodnala SK et al used an eGFP –mCherry-LC3b fusion reporter system to evaluate the impact of extracellular potassium concentration on autophagic flux in live T cells, using an autophagy-incompetent construct containing a Gly120 → Ala substitution (G120A) in LC3b as a negative control [20].

Another plasmid that has been recently developed to include an internal control is GFP-LC3-RFP-LC3ΔG. Upon cleavage of this protein by Atg4 in the cells, two distinct populations of LC3 are released, GFP-LC3 that decorates the autophagosomes and RFP-LC3ΔG that remains [38] in the cytosol and serves as an internal control for the quantity of GFP-LC3 before it is degraded by the autolysosomes. The outcome measure used in this assay is the ratio of GFP/RFP that is decreased upon an increase in autophagy.

Despite the certain advantages of LC3 puncta method, the galectin puncta formation assay is believed to be better for analyzing lysosomal membrane permeabilization due to its high sensitivity [24]. This technique employs the process of galectin translocation to impaired lysosomes, which is followed by a switch from homogenous to punctate staining distribution.

Flow cytometry

Detection of autophagosomes can also be monitored by using Flow cytometry. While microscopy is a great technique to visualize the autophagosomes in a small population of cells at a given time, flow cytometry offers a high-throughput analysis of the autophagosomes. Moreover, flow cytometry can also be used to detect autophagosomes in non-adherent cells such as blood monocytes. The outcome measured by flow cytometry is a decrease in GFP-LC3 fluorescence which is based on the principle that GFP-LC3 is degraded in the autolysosome and thus a decrease in GFP signal would indicate autophagy flux [39]. Flow cytometry can also be used with dyes such as CYTO-ID which are selectively accumulated in the autophagic vacuoles. CYTO-ID can also be detected by fluorescence microscopy [4].

Tandem labeled fluorescent LC3 expression vectors can also be analyzed with flow cytometry, concurrently with the fluorescent microscopy analysis as discussed above. Vodnala SK et al used the proportion of mCherry+GFP cells in flow cytometry as a parameter for autophagic flux [20].

P62 degradation

P62 is an adaptor receptor that recruits cargo to the autophagosome. P62 also binds to the LC3 on the autophagosomes. P62 is degraded along with the designated cargo in the autolysosomes [11]. Thus, p62 degradation is considered a biomarker for autophagy activity. However, caution must be taken as p62 is also degraded by autophagy-independent pathways such as proteasomes. Thus, to confirm autophagy-mediated p62 degradation, one must use lysosomal inhibitors such as bafilomycin A1 to demonstrate that p62 is no longer degraded in the presence of this inhibitor. E Gelpi et al proposed to use nuclear p62 staining to identify Alzheimer type II astrocytes in metabolic/hepatic encephalopathy [40].

Turnover of long-lived proteins

Traditionally this method involves labeling long-lived proteins using radiolabeled amino acids such as 35S-methionine 3H-leucine, 14C-leucine, and 14C-valine [41] before inducing autophagy and monitoring the release of radioactivity as a measure of degradation of long-lived proteins upon induction of autophagy [29]. Again, one should use autophagy inhibitors such as bafilomycin A1 to demonstrate that the degradation of long-lived protein is mediated by autophagy and not by proteasomes. A non-radioactive method of detecting long-lived protein turnover is by using L-azidohomoalanine (AHA) through click-chemistry [27, 42].

Transmission electron microscopy

Electron microscopy remains the best method for visualizing autophagosomes at the ultrastructural level. The nature of the specific cargo can also be detected using this technique. In addition, immunogold labeling can be utilized to identify special cargo and proteins within the autophagosomes [31]. While TEM remains by far the best technique to visualize autophagosomes, it presents significant challenges. The sample preparation is very critical when processing for EM and may require specific methods and expertise to maintain the integrity of the organelles. TEM is also a time-consuming technique and proper identification of the autophagic structure is difficult. Improper sample processing may also lead to the appearance of artifacts that may resemble autophagosomes [43]. Also, significant improvement in the visualizing of autophagosome ultrastructure has recently been achieved by implementing new fixation methods, such as ferrocyanide-reduced osmium and aldehyde/OsO4 mixture for detecting omegasome structures [30].

Autophagy gene and protein expression

Increase in the mRNA and protein expression of several autophagy core genes and proteins such as Atg7, Beclin1, Atg5, LC3 often accompany the induction of autophagy [32, 44-46]. These outcomes are measured by real-time qPCR or northern blotting for analyzing gene expression and by western blotting using specific antibodies to analyse the protein expression. However, changes in mRNA and protein levels of autophagy players may not always be observed upon induction of autophagy and should not be used as a sole criterion to demonstrate the activation of autophagy and must be accompanied by other assays demonstrating functional autophagy.

Keima

Keima is a coral-derived fluorescent protein with a bimodal excitation spectrum depending on the pH. Keima exhibits excitation at 440 (appears as green) and 586 nm (appears as red) in neutral and acidic environments, respectively. Keima exhibits a low 550/438 excitation ratio in the cytosol under basal conditions. Upon induction of autophagy, an increase in the punctate structures with a higher 550/438 ratio is observed which represent autolysosomes [33]. Keima provides advantages over the RFP-GFP-LC3 system as it does not depend on the LC3 lipidation system and is useful in detecting autophagy that is independent of Atg5 conjugation system. However, Keima cannot be used in the fixed cells as it requires the lysosomal acidity to function. Keima has been successfully used for demonstrating mitophagy.

Cargo sequestration assay

The cargo sequestration analysis applies a cargo marker of total cytplasm to measure bulk autophagy [34]. A recent study assessed the sequestration of cytosolic protein lactate dehydrogenase in autophagic vacuoles of cells treated with inhibitors of intravacuolar degradation. The method was based on electrodisruption of the cell membrane followed by centrifugal sedimentation.

Semiconductor-based in situ measurement

A recent study has presented a novel semiconductor-based method to estimate autophagy induced by nutrient starvation [35]. The authors have used a semiconductor-based field-effect transistor (FET) biosensor to monitor pH changes in starved HeLa cells. The detected positive shift correlated with the elevated numbers of hydrogen ions due to increased cellular respiration in autophagic cells.

Monitoring Autophagy in Animals

Assays such an LC3 turnover and p62 degradation by western blot and immunohistochemistry can also be monitored in animal tissues [47, 48]. However, the optimal detection of LC3 and p62 in animal tissues is difficult and may depend on the source of tissue and the method of protein extraction. Some tissues have a higher level of LC3-I while others show a much higher level of LC3-II. Moreover, tissues have a heterogeneous population of cells which may present challenge of variability in detecting LC3-II. Moreover, autophagy flux is difficult to study in the whole animal and require the administration of flux inhibitors such as bafilomycin which may be toxic to the animals. Under such circumstances, ex vivo methods may be utilized where tissues are isolated and treated with the inhibitors and autophagy flux is determined by western blotting. Transgenic mice expressing GFP-LC3 have also been utilized to monitor autophagy in vivo [47]. Typically, cryosections are used for detecting GFP-LC3 positive autophagosomes in such animal models for testing autophagy induction. Advancement of this model is the development of mice which express RFP-GFP-LC3 [49, 50]. Moreover, cells derived from such animals can also be grown in vitro and used for further analysis [51]. Recently, mice expressing GFP-LC3-RFP-LC3ΔG have also been developed [38]. Such mice models would be very valuable in studying autophagy induction in vivo. Conventional and organ-specific autophagy-deficient mouse models have also been fundamental in advancing our knowledge of autophagy [52].

Commercially Available Autophagy Detection Kits

In addition to conventionally used laboratory methods, commercially available kits from various sources are also available that maybe used in conjunction with the methods described above. These kits often use certain dyes to label the autophagic vacuoles which then can be detected by fluorescence microscopy, flow cytometry, or a microplate reader. Some of these kits are:

  • Autophagy Assay Kit (ab139484): Abcam
  • Autophagy Assay Kit (MAK138-1KT):Sigma Aldrich
  • CYTO-ID autophagy detection kit (ENZ-KIT175-0050): Enzo
  • Autophagy Detection Reagent Pack (CF200097): Millipore

Of these, CYTO-ID detection kit has been used relatively commonly by several researchers to monitor autophagy in their studies [4, 53]. The dye used in this kit is a cationic amphiphilic tracer (CAT) dye that has been titrated to a dose that specifically labels autophagic vacuoles. Typically cells are first treated with a test reagent. Use of positive and negative controls such as rapamycin and chloroquine respectively are highly recommended for this assay. After incubation, the cells are washed and incubated in the dark with the CYTO-ID detection reagent and cells are then and analyzed by microscopy or flow cytometry. Both adherent and suspension cells can be analyzed using this reagent. Unlike conventional laboratory autophagy detection assays, these assays are less time-consuming. However, these assays should always be used in conjunction with more standard methods of monitoring autophagy rather than being used as a single method to detect autophagy.

Conclusion

Field of autophagy is dynamic and is ever evolving. New assays and techniques are constantly being developed to overcome existing caveats and to make progress in our current understanding of this field. Today, several methods exist that assist in the detection of autophagy in vitro and an in vivo. However, each method presents its own significant strengths and weaknesses. Thus, it is highly recommended that researchers use multiple methods when demonstrating autophagy in their experimental models and use appropriate positive and negative controls, wherever possible. In addition, researchers should also measure autophagy flux in their assays as a demonstration of a complete autophagic pathway. Finally, the autophagic assays should also be validated using genetic or pharmacological inhibitors of autophagy.

References
  1. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147:728-41 pubmed publisher
  2. Yamamoto H, Zhang S, Mizushima N. Autophagy genes in biology and disease. Nat Rev Genet. 2023;24:382-400 pubmed publisher
  3. Boukhalfa A, Nascimbeni A, Ramel D, Dupont N, Hirsch E, Gayral S, et al. PI3KC2α-dependent and VPS34-independent generation of PI3P controls primary cilium-mediated autophagy in response to shear stress. Nat Commun. 2020;11:294 pubmed publisher
  4. Silva M, Nandi G, Tentarelli S, Gurrell I, Jamier T, Lucente D, et al. Prolonged tau clearance and stress vulnerability rescue by pharmacological activation of autophagy in tauopathy neurons. Nat Commun. 2020;11:3258 pubmed publisher
  5. Magupalli V, Negro R, Tian Y, Hauenstein A, Di Caprio G, Skillern W, et al. HDAC6 mediates an aggresome-like mechanism for NLRP3 and pyrin inflammasome activation. Science. 2020;369: pubmed publisher
  6. Li W, Li J, Bao J. Microautophagy: lesser-known self-eating. Cell Mol Life Sci. 2012;69:1125-36 pubmed publisher
  7. Cuervo A, Wong E. Chaperone-mediated autophagy: roles in disease and aging. Cell Res. 2014;24:92-104 pubmed publisher
  8. Cui Y, Parashar S, Zahoor M, Needham P, Mari M, Zhu M, et al. A COPII subunit acts with an autophagy receptor to target endoplasmic reticulum for degradation. Science. 2019;365:53-60 pubmed publisher
  9. Russell R, Tian Y, Yuan H, Park H, Chang Y, Kim J, et al. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat Cell Biol. 2013;15:741-50 pubmed publisher
  10. Ohsumi Y, Mizushima N. Two ubiquitin-like conjugation systems essential for autophagy. Semin Cell Dev Biol. 2004;15:231-6 pubmed
  11. Johansen T, Lamark T. Selective autophagy mediated by autophagic adapter proteins. Autophagy. 2011;7:279-96 pubmed
  12. Itakura E, Kishi Itakura C, Mizushima N. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell. 2012;151:1256-69 pubmed publisher
  13. Chávez M, Morales R, Lopez Crisosto C, Roa J, Allende M, Lavandero S. Autophagy Activation in Zebrafish Heart Regeneration. Sci Rep. 2020;10:2191 pubmed publisher
  14. Saito T, Kuma A, Sugiura Y, Ichimura Y, Obata M, Kitamura H, et al. Autophagy regulates lipid metabolism through selective turnover of NCoR1. Nat Commun. 2019;10:1567 pubmed publisher
  15. Johmura Y, Yamanaka T, Omori S, Wang T, Sugiura Y, Matsumoto M, et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science. 2021;371:265-270 pubmed publisher
  16. Palla A, Ravichandran M, Wang Y, Alexandrova L, Yang A, Kraft P, et al. Inhibition of prostaglandin-degrading enzyme 15-PGDH rejuvenates aged muscle mass and strength. Science. 2020;: pubmed publisher
  17. Ling Q, Broad W, Trösch R, Töpel M, Demiral Sert T, Lymperopoulos P, et al. Ubiquitin-dependent chloroplast-associated protein degradation in plants. Science. 2019;363: pubmed publisher
  18. Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy. 2007;3:542-5 pubmed
  19. Tanida I, Minematsu Ikeguchi N, Ueno T, Kominami E. Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy. 2005;1:84-91 pubmed
  20. Vodnala S, Eil R, Kishton R, Sukumar M, Yamamoto T, Ha N, et al. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science. 2019;363: pubmed publisher
  21. Jiang P, Mizushima N. LC3- and p62-based biochemical methods for the analysis of autophagy progression in mammalian cells. Methods. 2015;75:13-8 pubmed publisher
  22. Maniatis S, Aijö T, Vicković S, Braine C, Kang K, Mollbrink A, et al. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science. 2019;364:89-93 pubmed publisher
  23. Tamaki Y, Shodai A, Morimura T, Hikiami R, Minamiyama S, Ayaki T, et al. Elimination of TDP-43 inclusions linked to amyotrophic lateral sclerosis by a misfolding-specific intrabody with dual proteolytic signals. Sci Rep. 2018;8:6030 pubmed publisher
  24. Aits S. Methods to Detect Loss of Lysosomal Membrane Integrity. Methods Mol Biol. 2019;1880:315-329 pubmed publisher
  25. Lee J, Yang D, Goulbourne C, Im E, Stavrides P, Pensalfini A, et al. Faulty autolysosome acidification in Alzheimer's disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat Neurosci. 2022;: pubmed publisher
  26. Hwang S, Han B, Lee M. Knockout of ATG5 leads to malignant cell transformation and resistance to Src family kinase inhibitor PP2. J Cell Physiol. 2018;233:506-515 pubmed publisher
  27. Li Z, Wang C, Wang Z, Zhu C, Li J, Sha T, et al. Allele-selective lowering of mutant HTT protein by HTT-LC3 linker compounds. Nature. 2019;: pubmed publisher
  28. SenthilKumar G, Skiba J, Kimple R. High-throughput quantitative detection of basal autophagy and autophagic flux using image cytometry. Biotechniques. 2019;67:70-73 pubmed publisher
  29. Dupont N, Leroy C, Hamai A, Codogno P. Long-Lived Protein Degradation During Autophagy. Methods Enzymol. 2017;588:31-40 pubmed publisher
  30. Arai R, Waguri S. Improved Electron Microscopy Fixation Methods for Tracking Autophagy-Associated Membranes in Cultured Mammalian Cells. Methods Mol Biol. 2019;1880:211-221 pubmed publisher
  31. Lucocq J, Hacker C. Cutting a fine figure: On the use of thin sections in electron microscopy to quantify autophagy. Autophagy. 2013;9:1443-8 pubmed publisher
  32. Nara A, Mizushima N, Yamamoto A, Kabeya Y, Ohsumi Y, Yoshimori T. SKD1 AAA ATPase-dependent endosomal transport is involved in autolysosome formation. Cell Struct Funct. 2002;27:29-37 pubmed
  33. Katayama H, Kogure T, Mizushima N, Yoshimori T, Miyawaki A. A sensitive and quantitative technique for detecting autophagic events based on lysosomal delivery. Chem Biol. 2011;18:1042-52 pubmed publisher
  34. Engedal N, Luhr M, Szalai P, Seglen P. Measurement of Bulk Autophagy by a Cargo Sequestration Assay. Methods Mol Biol. 2019;1880:307-313 pubmed publisher
  35. Sakata T, Saito A, Sugimoto H. In situ measurement of autophagy under nutrient starvation based on interfacial pH sensing. Sci Rep. 2018;8:8282 pubmed publisher
  36. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000;19:5720-8 pubmed
  37. Kimura S, Noda T, Yoshimori T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy. 2007;3:452-60 pubmed
  38. Kaizuka T, Morishita H, Hama Y, Tsukamoto S, Matsui T, Toyota Y, et al. An Autophagic Flux Probe that Releases an Internal Control. Mol Cell. 2016;64:835-849 pubmed publisher
  39. Shvets E, Fass E, Elazar Z. Utilizing flow cytometry to monitor autophagy in living mammalian cells. Autophagy. 2008;4:621-8 pubmed
  40. Gelpi E, Rahimi J, Klotz S, Schmid S, Ricken G, Forcen Vega S, et al. The autophagic marker p62 highlights Alzheimer type II astrocytes in metabolic/hepatic encephalopathy. Neuropathology. 2020;: pubmed publisher
  41. Bauvy C, Meijer A, Codogno P. Assaying of autophagic protein degradation. Methods Enzymol. 2009;452:47-61 pubmed publisher
  42. Wang J, Zhang J, Lee Y, Ng S, Shi Y, Hua Z, et al. Nonradioactive quantification of autophagic protein degradation with L-azidohomoalanine labeling. Nat Protoc. 2017;12:279-288 pubmed publisher
  43. Eskelinen E. To be or not to be? Examples of incorrect identification of autophagic compartments in conventional transmission electron microscopy of mammalian cells. Autophagy. 2008;4:257-60 pubmed
  44. Bernard A, Jin M, González Rodríguez P, Füllgrabe J, Delorme Axford E, Backues S, et al. Rph1/KDM4 mediates nutrient-limitation signaling that leads to the transcriptional induction of autophagy. Curr Biol. 2015;25:546-55 pubmed publisher
  45. Kovsan J, Bluher M, Tarnovscki T, Kloting N, Kirshtein B, Madar L, et al. Altered autophagy in human adipose tissues in obesity. J Clin Endocrinol Metab. 2011;96:E268-77 pubmed publisher
  46. Vazquez P, Arroba A, Cecconi F, de la Rosa E, Boya P, de Pablo F. Atg5 and Ambra1 differentially modulate neurogenesis in neural stem cells. Autophagy. 2012;8:187-99 pubmed publisher
  47. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 2004;15:1101-11 pubmed
  48. Waguri S, Komatsu M. Biochemical and morphological detection of inclusion bodies in autophagy-deficient mice. Methods Enzymol. 2009;453:181-96 pubmed publisher
  49. Hariharan N, Zhai P, Sadoshima J. Oxidative stress stimulates autophagic flux during ischemia/reperfusion. Antioxid Redox Signal. 2011;14:2179-90 pubmed publisher
  50. Castillo K, Valenzuela V, Matus S, Nassif M, Onate M, Fuentealba Y, et al. Measurement of autophagy flux in the nervous system in vivo. Cell Death Dis. 2013;4:e917 pubmed publisher
  51. Li L, Wang Z, Hill J, Lin F. New autophagy reporter mice reveal dynamics of proximal tubular autophagy. J Am Soc Nephrol. 2014;25:305-15 pubmed publisher
  52. Kuma A, Komatsu M, Mizushima N. Autophagy-monitoring and autophagy-deficient mice. Autophagy. 2017;13:1619-1628 pubmed publisher
  53. Xie S, García Prat L, Voisin V, Ferrari R, Gan O, Wagenblast E, et al. Sphingolipid Modulation Activates Proteostasis Programs to Govern Human Hematopoietic Stem Cell Self-Renewal. Cell Stem Cell. 2019;25:639-653.e7 pubmed publisher
ISSN : 2329-5139