Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1226-7155(Print)
ISSN : 2287-6618(Online)
International Journal of Oral Biology Vol.50 No.3 pp.103-111
DOI : https://doi.org/10.11620/IJOB.2025.50.3.103

Sesamin isolated from Hypericum hookerianum induces autophagy activation

Qui Ngoc Sang Nguyen1,2, Heesu Lee2, Jae Wook Lee1,3*
1Gangneung Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
2College of Dentistry, Gangneung Wonju National University, Gangneung 25457, Republic of Korea
3Natural Product Apply Science, KIST School, University of Science and Technology, Gangneung 25451, Republic of Korea
*Correspondence to: Jae Wook Lee, E-mail: jwlee5@kist.re.krhttps://orcid.org/0000-0002-0171-2160
June 19, 2025 August 22, 2025 August 23, 2025

Abstract


Autophagy is a ubiquitous and fundamental catabolic vital process for maintaining cellular homeostasis, achieved by degrading and recycling cytoplasmic components, particularly under conditions of nutrient deprivation or metabolic stress. This mechanism is also integral to the selective clearance of misfolded or aggregated proteins, the removal of dysfunctional organelles (such as mitochondria and the endoplasmic reticulum), and the intracellular degradation of pathogens, including those associated with peroxisomes. In this study, we screened and identified sesamin, a bioactive compound isolated from Hypericum hookerianum extracts, as a novel autophagy activator. Our results demonstrated that sesamin effectively induces autophagy and activates the lysosome biogenesis pathway.



초록


    © The Korean Academy of Oral Biology. All rights reserved.

    This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/bync/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

    Introduction

    Autophagy is a cellular process in which the cell degrades and recycles its components to maintain homeostasis. As a fundamental catabolic process, autophagy involves the lysosome-mediated degradation of damaged, unnecessary, or dysfunctional cellular components [1,2]. Autophagy also plays a critical role in cellular metabolism and in regulating cell death and survival [3,4]. Under normal physiological conditions, most eukaryotic cells exhibit basal autophagy. In contrast, autophagy-deficient cells display extensive abnormal protein accumulation and mitochondrial disorganization, indicating that autophagy plays a critical role in the clearance of damaged organelles and protein aggregates that are resistant to degradation by the ubiquitin-proteasome system, thereby contributing to the maintenance of cellular homeostasis [5,6]. Genetic and mechanistic studies have highlighted autophagy’s involvement in various human health conditions, including cancer [7,8], neurodegenerative diseases [9,10], age-related disorders, and inflammatory and infectious diseases [11,12].

    Hypericum hookerianum Wight & Arnott (family Hypericaceae), widely allocated across Asian countries such as China, Bhutan, India, Myanmar, Nepal, Thailand, and Vietnam [13], is well known in folklore medicine for its diverse therapeutic properties, including antioxidant [14,15], anticancer [16,17], antidepressants [18,19], neuroprotection [20], and antifungal activities [21-23]. In this study, we investigated the impact of sesamin on the activation of autophagy.

    Materials and Methods

    1. Cell culture

    The HeLa and cells were obtained from the Korea Cell Line Bank and cultured in high-glucose Dulbecco’s modified Eagle’s medium supplemented with 10% heat-inactivated fetal bovine serum and 1% penicillin/streptomycin. Cells were maintained at 37℃ in a humidified incubator with 5% CO2.

    2. Autophagy screening method

    The plasmid pMRX-IP-GFP-LC3-RFP-LC3ΔG, containing an autophagic flux probe with an internal control, was obtained from Addgene (Cat. No. 84572). The HeLa cell line expressing this construct was designated as HeLa-LC3BΔG. Cells were seeded at a density of 30,000 HeLa-LC3BΔG cells per well in a black-walled, clear-bottom 96-well plate (Greiner). After 24 hours of incubation, cells were treated with Sesamin and a positive control. Following a 24-hour treatment period, cells were fixed with 4% paraformaldehyde for 10 minutes and washed with Dulbecco’s phosphate-buffered saline (DPBS). Fluorescence intensities for green fluorescent protein (GFP) (488 nm ± 9, 514 nm ± 15) and red fluorescent protein (RFP) (584 nm ± 9, 612 nm ± 15) were measured using a Tecan Infinite 1000 Series plate reader (Tecan).

    The HEK293-LC3-Hibit cell was provided by Promega (Cat. No. GA1040). Cells were seeded at a density of 20,000 HEK293-LC3-Hibit cells per well in a white-walled, flat-bottom 96-well plate (Greiner). After 18 hours of incubation, cells were treated with sesamin and a positive control. Following a 24-hour treatment period, the nano buffer was added for each well, and the luminescent intensity was measured by the Glomax machine (Promega).

    3. Acridine orange staining

    HeLa cells were seeded in a black-walled, clear-bottom 96- well plate at a density of 5,000 cells per well. After 12 hours of incubation, cells were treated with sesamin for 24 hours. Following treatment, the culture medium was removed, and the wells were washed with DPBS. Cells were then stained with 2 µM acridine orange for 15 minutes, followed by another DPBS wash before being photographed using the Operetta high-content imaging system (Perkin Elmer). The fluorescence intensity was estimated using the Harmony 3.5 program (Perkin Elmer).

    4. Lysosome pH measurement

    HeLa cells were seeded at a density of 8,000 cells per well in a black-walled, clear-bottom 96-well plate. After overnight incubation, the culture medium was replaced, and cells were treated with sesamin 50 µM, rapamycin 25 µM, bafilomycin A1 20 nM, or a combination of sesamin 50 µM and bafilomycin A1 20 nM. After 24 hours of incubation, half of the culture medium was removed, and 50 µL of 2 µM DND-160 (L7545; Thermo Fisher Scientific) prepared in fresh medium was added to each well. Cells were incubated for 15 minutes and subsequently washed with DPBS. Fluorescence was measured at excitation/ emission wavelengths of 329/540 nm (yellow) and 329/440 nm (blue), with a bandwidth of 10 nm, using the Tecan Infinite 1000 Series plate reader. Imaging was performed using the Operetta high-content imaging system. The fluorescence intensity was estimated using the Harmony 3.5 program.

    5. Immunoblot analysis

    HeLa cells (2 × 105) were seeded in a 6-well plate (Corning) and incubated overnight. After overnight growth, the cells were treated with compounds, and after 24 hours, the cells were harvested. First, cells were lysed using cell extraction buffer (Invitrogen) along with a complete protease inhibitor cocktail (Sigma-Aldrich) and phenylmethanesulfonyl fluoride (1 mM). Cells were lysed for 30 minutes and spun down at 14,000 rpm for 20 minutes. The supernatant containing total protein was quantified using the BCA protein assay kit (Invitrogen). Furthermore, a total of 10 µg of protein was loaded onto an SDS-PAGE gel and electrophoresed at 100 V for 120 minutes. Proteins were then transferred onto a polyvinylidene fluoride membrane (Millipore) at 100 V for 90 minutes. Membranes were blocked with either 5% skimmed milk or 3% bovine serum albumin (for phosphorylated proteins) for 1 hour at room temperature. Primary antibodies (1:1,000 dilution) were incubated overnight at 4℃, followed by incubation with secondary antibodies (1:1,000 dilution) at room temperature for 90 minutes. Chemiluminescent signals were detected using the SuperSignal West Pico Plus reagent (Thermo Fisher Scientific), and images were captured.

    6. Data analysis

    All graphs were generated using GraphPad Prism 10 software (GraphPad). Three independent experiments present data as mean ± standard deviation. Statistical significance was determined using one-way nonparametric analysis of variance.

    Results

    1. Identification of sesamin- an autophagy activator from H. hookerianum

    To characterize the autophagy-promoting activity of individual constituents isolated from H. hookerianum, we subjected all 28 purified compounds to the puncta-formation assay (Fig. 1A and 1B, Supplementary Table 1, Supplementary Fig. 1). Autophagy activation is traditionally assessed by monitoring the turnover of lipidated LC3B (LC3B-II) via immunoblotting. While effective, this method is labor-intensive, time-consuming, and resource-demanding. As a result, cell-based reporter assays utilizing fluorescently tagged LC3B have emerged as a more practical alternative for high-throughput screening. Initial systems employed GFP-LC3B constructs to visualize autophagosome formation, as GFP-LC3B is incorporated into autophagosomal membranes upon induction of autophagy. However, the GFP signal is quenched in the acidic environment of autolysosomes, limiting the ability to monitor autophagic flux. To address this limitation, dual-tagged RFP-GFP-LC3B constructs were developed. While this system allows differentiation between autophagosomes (GFP+/RFP+) and autolysosomes (GFP–/RFP+), it does not quantitatively assess autophagic flux due to the persistence of non-lipidated LC3 forms. To overcome these constraints, the GFP-LC3B-RFP-LC3ΔG reporter assay was established [1,2]. In the GFP-LC3B-RFP-LC3ΔG reporter assay, GFP is fused to full-length LC3B, whereas RFP is fused to an LC3B variant lacking the N-terminal glycine residue (LC3ΔG). Upon induction of autophagy, the cysteine protease ATG4 cleaves full-length LC3B at its N-terminus, releasing GFP-LC3B and RFP-LC3ΔG [24,25]. The liberated GFP-LC3B conjugates to phosphatidylcholine and is incorporated into autophagosomal membranes, ultimately undergoing lysosomal degradation. In contrast, RFP-LC3ΔG lacks the essential glycine required for lipidation and remains cytosolic. Consequently, the ratio of GFP to RFP fluorescence provides a quantitative measure of autophagic flux, with a decrease in the GFP/RFP signal ratio reflecting increased autophagic degradation. This system enables more accurate and dynamic measurement of autophagic flux by distinguishing between functional autophagy and static autophagosome accumulation. HeLa-GFP-LC3B-RFP-LC3ΔG cells were incubated with either vehicle (dimethyl sulfoxide [DMSO]) or 50 µM of each compound for 24 hours. Following treatment, cells were fixed, and automated high-content imaging was performed on the Operetta platform.

    Quantitative image analysis revealed that sesamin (HH28), which was isolated from dichloromethane extract, has been previously reported for its neuroprotective effects. This result identifies sesamin as a potent activator of autophagy among the 28 H. hookerianum’s single compounds.

    2. Sesamin activates autophagy and autophagic flux

    Autophagy activation leads to the formation of autophagosome vesicles, which sequester unwanted cellular components for degradation. LC3B is predominantly recruited to the inner membrane of autophagosomes. Therefore, HeLa-GFP-LC3B-RFP-LC3ΔG cells were utilized to examine autophagic vesicle formation. Sesamin treatment induces a significant decline in the GFP/RFP ratio and a marked increase in LC3B puncta, indicating the formation of autolysosomes (Fig. 2B2D, Supplementary Fig. 2). Torin-1 and bafilomycin A1 were used as an autophagy activator and inhibitor, respectively. Furthermore, exposure to sesamin led to a decreased luminescent signal in the Autophagy-LC3B-HiBiT assay (Fig. 2E). Upon autophagy activation, LC3BI undergoes processing and conversion to LC3BII, which is subsequently recruited to the inner membrane of the autophagosome. Therefore, the LC3BII/LC- 3BI ratio serves as a marker of autophagy. As illustrated in the figure, sesamin increased the ratio of LC3BII/LC3BI levels as determined by immunoblotting (Fig. 3A and 3B). On the other hand, p62 facilitates the delivery of ubiquitinated cargo for autophagic degradation through its C-terminal ubiquitin-associated or LC3-interacting region domain [26]. Consequently, p62 levels increase during the early stages of autophagy, but it is degraded along with its cargo inside the autolysosome. Therefore, p62 is widely used as a marker of autophagic flux. The p62 expression increases after 6 hours treatment with sesamin, decreases after 12 hours, and disappears completely after 24 hours, indicating sesamin-induced autophagic flux (Fig. 3C and 3D).

    3. Sesamin activates lysosome biogenesis

    Autophagic flux during starvation is facilitated by increased lysosomal activity. Perinuclear lysosomes exhibit a more acidic environment, enhancing enzymatic function and promoting degradation [27]. For this reason, the acridine orange was used to determine the lysosome generation. The red color of acridine orange increases at an acidic pH and is used to describe acidic lysosomal formation. Sesamin treatment for 24 hours significantly increased acid-red fluorescent in a dosedependent manner, indicating that sesamin induces lysosome formation (Fig. 4A and 4B). Meanwhile, lysosomal pH was analyzed in HeLa cells following incubation with DMSO, 20 nM bafilomycin A1, 25 μM rapamycin, and 50 μM sesamin for 24 hours, after that the cell was stained by LysoSensor DND-160 for 15 minutes. A reduced yellow-to-blue ratio is indicative of increased pH. The treatment with sesamin induces an increase in the ratio of yellow-to-blue, indicating the pH reduction. However, treatment with bafilomycin A1, an autophagy inhibitor, decreases the yellow-to-blue ratio, indicating increased pH (Fig. 5A and 5B).

    4. Sesamin shows autophagic neuroprotection against oxidative stress

    In this study, the neuroprotective effect of sesamin against oxidative stress-induced neuronal cell death via autophagy activation was investigated. HT22 cells were treated with sesamin in the presence or absence of the autophagy inhibitor (chloroquine [CQ]), followed by glutamate-induced oxidative stress. Sesamin treatment significantly protected neuronal cells, whereas co-treatment with CQ abolished this effect. These results indicate that sesamin exerts its neuroprotective activity through autophagy activation under oxidative stress conditions (Fig. 6A and 6B).

    Discussion

    This study identified sesamin, a bioactive compound isolated from H. hookerianum , as a potent autophagy activator. Our findings demonstrate that sesamin promotes autophagy by increasing the LC3B-II/LC3B-I ratio and enhancing autophagic flux, which are hallmarks of effective autophagy induction.

    Activation of lysosomal biogenesis by sesamin further strengthens its role as an autophagy enhancer. Lysosomes play a critical role in the final degradation of autophagic cargo, and their impairment is commonly observed in neurodegenerative diseases [27]. Our study demonstrates that sesamin increases lysosomal acidification, as evidenced by enhanced acridine orange staining and LysoSensor assays, indicating improved lysosomal function. This aligns with previous research suggesting that stimulating lysosomal activity can enhance the clearance of toxic protein aggregates, further underscoring sesamin’s potential in neuroprotection.

    While our study provides strong evidence for the role of sesamin in autophagy activation further investigations are needed to elucidate its precise molecular mechanisms. Specifically, identifying the direct molecular targets of sesamin in the autophagy-lysosomal pathway will provide deeper insights into its mode of action. Additionally, in vivo studies using animal models will be crucial to validate our in vitro findings and assess the potential therapeutic application of sesamin.

    Sesamin, isolated from H. hookerianum, is a potential autophagy activator. Sesamin significantly increased the autophagosome and reduced pH levels in HeLa cells. Moreover, sesamin promoted autophagic flux, as evidenced by increased LC3B-II and changed p62 expression time-dependent, along with enhanced lysosomal activity. These findings highlight sesamin as a promising natural compound for therapeutic strategies aimed at autophagy-related diseases. In vivo studies and further mechanistic investigations are needed to confirm the potential of sesamin for clinical application in autophagyrelated diseases.

    Funding

    KIST internal grant (2G13220).

    Acknowledgements

    We sincerely thank Dr. Phuong Thien Thuong, VKIST for providing the materials for this study.

    Conflicts of Interest

    No potential conflict of interest relevant to this article was reported.

    Supplementary Data

    Supplementary data is available at http://www.kijob.or.kr only.

    Figure

    IJOB-50-3-103_F1.jpg

    Found out sesamin induces autophagy from Hypericum hookerianum extract. (A) H. hookeriaum single compounds’ image analysis induces autophagosome vesicle formation in HeLa-GFP-LC3B-RFP-LC3ΔG. (B) The bar graph was generated based on the puncta image. The data’s bar graphs are presented as means ± standard deviation of n = 3.

    DMSO, dimethyl sulfoxide.

    **p < 0.02, ***p < 0.001 compared with DMSO.

    IJOB-50-3-103_F2.jpg

    Sesamin induces autophagy. (A) The structure of sesamin. (B) Sesamin, Torin-1, and bafilomycin A1 were treated in HeLa-GFP-LC3B-RFP-LC3ΔG. After 24 hours incubation, the fluorescence intensity was assessed at RFP (584 nm ± 9, 612 nm ± 15), GFP (488 nm ± 9, 514 nm ± 15), and the ratio GFP/ RFP was calculated. (C) The bar graph shows the effect of sesamin-inducing autophagosome vesicle formation. (D) The image analysis shows the effect of sesamin-inducing autophagosome vesicle formation. Sesamin and Torin-1 were treated in HeLa-GFP-LC3B-RFP-LC3ΔG. After 24 hours incubation, the cells were taken image using Operetta (scale bar: 20 μm) and analyzed using Harmony 3.5 software. (E) Autophagy activation of sesamin using HEK293-LC3- Hibit assay. Four extracts of Hypericum hookeriaum , Torin-1, rapamycin, and bafilomycin A1 were treated in HEK293-LC3-Hibit cells. After 24 hours incubation, the luminescent intensity was read out. The data’s bar graphs are presented as means ± standard deviation of n = 3.

    GFP, green fluorescent protein; RFP, red fluorescent protein; DMSO, dimethyl sulfoxide.

    ***p < 0.001 compared with DMSO.

    IJOB-50-3-103_F3.jpg

    (A) Western blot was conducted to examine the protein expression of LC3B using sesamin. (B) The bar graphs were generated from Western blot data. (C) Sesamin induces p62 expression in time-dependent. Sesamin treatment increases the p62 expression from 0–6 hours and significantly decreases after that. (D) Bar graphs representing the intensity of immunoblot bands, as quantified using Image J software (version 1.54k). The data’s bar graphs are presented as means ± standard deviation of n = 3.

    DMSO, dimethyl sulfoxide.

    **p < 0.003, ***p < 0.001 compared with DMSO.

    IJOB-50-3-103_F4.jpg

    Sesamin activates lysosome biogenesis. (A) The image analysis shows the effect of sesamin-inducing lysosome formation. Sesamin, CQ, and bafilomycin A1 were treated in HeLa cells. After 24 hours incubation, the cell was stained by Acridine orange dye for 15 minutes and the fluorescent intensity was read out by the Operetta machine and analyzed by Harmony 3.5 software. (B) The bar graph shows the effect of sesamin on inducing lysosome formation. The data’s bar graphs are presented as means ± standard deviation of n = 3.

    CQ, chloroquine; DMSO, dimethyl sulfoxide; AO, acridine orange.

    *p < 0.02, ***p < 0.001 compared with DMSO.

    IJOB-50-3-103_F5.jpg

    (A) The image analysis presents the effect of sesamin in reducing the lysosomal pH using LysoSensor DND-160, indicating the lysosome activation. (B) The bar graph shows the lysosome pH measurement. The data’s bar graphs are presented as means ± standard deviation of n = 3.

    DMSO, dimethyl sulfoxide.

    ***p < 0.001 compared with DMSO.

    IJOB-50-3-103_F6.jpg

    Neuroprotective effects of sesamin. (A) Autophagic neuroprotection of sesamin in HT22 cells. (B) Neuroprotective effects of sesamin in RGC5 cells. The data’s bar graphs are presented as means ± standard deviation of n = 3.

    CQ, chloroquine; DMSO, dimethyl sulfoxide; NAC, N-acetyl cysteine.

    *p < 0.06, **p < 0.005, ***p < 0.001 compared with DMSO.

    Table

    Reference

    1. Chung CY, Shin HR, Berdan CA, Ford B, Ward CC, Olzmann JA, Zoncu R, Nomura DK. Covalent targeting of the vacuolar H+-ATPase activates autophagy via mTORC1 inhibition. Nat Chem Biol 2019;15:776-85.
    2. Kaizuka T, Morishita H, Hama Y, Tsukamoto S, Matsui T, Toyota Y, Kodama A, Ishihara T, Mizushima T, Mizushima N. An autophagic flux probe that releases an internal control. Mol Cell 2016;64:835-49.
    3. Settembre C, Ballabio A. Lysosome: regulator of lipid degradation pathways. Trends Cell Biol 2014;24:743-50.
    4. Rabinowitz JD, White E. Autophagy and metabolism. Science 2010;330:1344-8.
    5. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006;441:885-9.
    6. Zhou Y, Manghwar H, Hu W, Liu F. Degradation mechanism of autophagy-related proteins and research progress. Int J Mol Sci 2022;23:7301.
    7. White E. The role for autophagy in cancer. J Clin Invest 2015; 125:42-6.
    8. Mulcahy Levy JM, Thorburn A. Autophagy in cancer: moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ 2020;27:843-57.
    9. Frake RA, Ricketts T, Menzies FM, Rubinsztein DC. Autophagy and neurodegeneration. J Clin Invest 2015;125:65-74.
    10. Menzies FM, Fleming A, Rubinsztein DC. Compromised autophagy and neurodegenerative diseases. Nat Rev Neurosci 2015;16:345-57.
    11. Deretic V, Kimura T, Timmins G, Moseley P, Chauhan S, Mandell M. Immunologic manifestations of autophagy. J Clin Invest 2015;125:75-84.
    12. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature 2011;469:323-35.
    13. Wahile A, Mukherjee K, Kumar V, Saha BP, Mukherjee PK. Antioxidant potentials of Hypericum hookerianum (family: Hypericaceae) on CCl4 induced hepatotoxicity in rats. Orient Pharm Exp Med 2007;7:85-93.
    14. Manosroi J, Wilairat R, Kijjoa A, Manosroi A. Free radical scavenging activity of extracts from Thai plants in Guttiferae and Schisandraceae families. Pharm Biol 2005;43:324-9.
    15. Pradeep PS, Manisha S, Monica Amala Nayaki J, Sivaraman D, Selvaraj R, Seeni S. Potential antioxidant and anti-inflammatory action of Hypericum hookerianum extracts in a liposome system evaluated with zebrafish embryos. J Microencapsul 2019;36:513-22.
    16. Dongre SH, Badami S, Natesan S, H RC. Antitumor activity of the methanol extract of Hypericum hookerianum stem against Ehrlich ascites carcinoma in Swiss albino mice. J Pharmacol Sci 2007;103:354-9.
    17. Dongre SH, Badami S, Godavarthi A. Antitumor activity of Hypericum hookerianum against DLA induced tumor in mice and its possible mechanism of action. Phytother Res 2008;22:23- 9.
    18. Sooriamuthu S, Varghese RJ, Bayyapureddy A, John SST, Narayanan R. Light-induced production of antidepressant compounds in etiolated shoot cultures of Hypericum hookerianum Wight & Arn. (Hypericaceae). Plant Cell Tiss Organ Cult 2013;115:169-78.
    19. Subakanmani S, Murugan S, Devi PU. Evaluation of antidepressant like effects of ethanolic Hypericum hookerianum and its glycosidic flavonoid enriched extract in reserpine induced Swiss albino mice. Asian J Biochem 2016;11:1-13.
    20. Hong VD, Vu VT, Selvaraj B, Tuyen NM, Khoi NM, Thuan ND, Thuong PT, Lee JW. Chemical constituents and neuroprotective activity of Hypericum hookerianum. Nat Prod Commun 2023;18.
    21. Wilairat R, Manosroi J, Manosroi A, Kijjoa A, Nascimento MS, Pinto M, Silva AM, Eaton G, Herz W. Cytotoxicities of xanthones and cinnamate esters from Hypericum hookerianum. Planta Med 2005;71:680-2.
    22. Wang QQ, Wang XD, Wu LZ, Fang QQ, Liu YN, Jiang K, Qu SJ, Tan CH. Polyprenylated acylphloroglucinols as deubiquitinating protease USP7 inhibitors from Hypericum hookerianum. Fitoterapia 2020;146:104678.
    23. Zhao J, Liu W, Wang JC. Recent advances regarding constituents and bioactivities of plants from the genus Hypericum. Chem Biodivers 2015;12:309-49.
    24. Tanida I, Sou YS, Ezaki J, Minematsu-Ikeguchi N, Ueno T, Kominami E. HsAtg4B/HsApg4B/autophagin-1 cleaves the carboxyl termini of three human Atg8 homologues and delipidates microtubule-associated protein light chain 3- and GABAA receptor-associated protein-phospholipid conjugates. J Biol Chem 2004;279:36268-76.
    25. Kabeya Y, Mizushima N, Yamamoto A, Oshitani-Okamoto S, Ohsumi Y, Yoshimori T. LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation. J Cell Sci 2004;117(Pt 13):2805-12.
    26. Liu WJ, Ye L, Huang WF, Guo LJ, Xu ZG, Wu HL, Yang C, Liu HF. p62 links the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell Mol Biol Lett 2016;21:29.
    27. Yim WW, Mizushima N. Lysosome biology in autophagy. Cell Discov 2020;6:6.