AutophagyPub Date : 2025-10-16DOI: 10.1080/15548627.2025.2576613
Einar K Krogsaeter, Justin McKetney, Leopoldo Valiente-Banuet, Angelica Marquez, Alexandra Willis, Zeynep Cakir, Erica Stevenson, Gwendolyn M Jang, Antara Rao, Emmy Li, Anton Zhou, Anjani Attili, Timothy S Chang, Martin Kampmann, Yadong Huang, Nevan J Krogan, Danielle L Swaney
{"title":"Lysosomal proteomics reveals mechanisms of neuronal APOE4-associated lysosomal dysfunction.","authors":"Einar K Krogsaeter, Justin McKetney, Leopoldo Valiente-Banuet, Angelica Marquez, Alexandra Willis, Zeynep Cakir, Erica Stevenson, Gwendolyn M Jang, Antara Rao, Emmy Li, Anton Zhou, Anjani Attili, Timothy S Chang, Martin Kampmann, Yadong Huang, Nevan J Krogan, Danielle L Swaney","doi":"10.1080/15548627.2025.2576613","DOIUrl":"https://doi.org/10.1080/15548627.2025.2576613","url":null,"abstract":"<p><p>APOE4 is the primary risk factor for Alzheimer disease (AD). Early AD pathological events first affect the neuronal endolysosomal system, which in turn causes neuronal protein aggregation and cell death. Despite the crucial influence of lysosomes upon AD pathophysiology, and that APOE4 localizes to lysosomes, the influence of APOE4 on lysosomal function remains unexplored. We find that expression of APOE4 in neuronal cell lines results in lysosomal alkalinization and impaired lysosomal function. To identify driving factors for these defects, we performed quantitative lysosomal proteome profiling. This revealed that APOE4 expression results in differential regulation of numerous lysosomal proteins, correlating with APOE allele status and disease severity in AD brains. In particular, APOE4 expression results in the depletion of lysosomal LGALS3BP and the accumulation of lysosomal TMED5. We additionally validated that these lysosomal protein changes can be targeted to modulate lysosomal function. Taken together, this work thereby reveals that APOE4 causes widespread lysosomal defects through remodeling the lysosomal proteome, with the lysosomal TMED5 accumulation and LGALS3BP depletion manifesting as lysosomal alkalinization in APOE4 neurons.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":""},"PeriodicalIF":14.3,"publicationDate":"2025-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145310285","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-16DOI: 10.1080/15548627.2025.2576615
Michele Di Crosta, Francesca Chiara Ragone, Rossella Benedetti, Gabriella D'Orazi, Roberta Santarelli, Roberta Gonnella, Maria Saveria Gilardini Montani, Mara Cirone
{"title":"Acetylation promotes mutant (MUT) TP53-HSPA8 and HSPA8-BAG3 interactions, facilitating MUT TP53 lysosomal degradation preferentially via CASA.","authors":"Michele Di Crosta, Francesca Chiara Ragone, Rossella Benedetti, Gabriella D'Orazi, Roberta Santarelli, Roberta Gonnella, Maria Saveria Gilardini Montani, Mara Cirone","doi":"10.1080/15548627.2025.2576615","DOIUrl":"https://doi.org/10.1080/15548627.2025.2576615","url":null,"abstract":"<p><p>Targeting mutant (MUT) TP53 is crucial in anticancer therapy, given the oncogenic properties that these proteins often acquire. Therefore, it is of paramount importance to unravel strategies and mechanisms through which this goal can be achieved. Valproic acid (VPA) downregulates the expression of MUT TP53 in several tumor cells, although the mechanisms involved remain to be explored. Here, we demonstrate for the first time that acetylation induced by VPA promotes the lysosomal degradation of MUT TP53 and that it occurs preferentially through chaperone-assisted selective autophagy (CASA). Indeed, acetylation of MUT TP53 increases its interaction with STUB1 (STIP1 homology and U-box containing protein 1), HSPB8 (heat shock protein family B (small) member 8) and HSPA8 (heat shock protein family A (Hsp70) member 8) and the latter, itself acetylated by VPA, binds to BAG3 (BAG cochaperone 3), facilitating the recruitment of MUT TP53 into the CASA pathway. These findings elucidate the mechanisms through which acetylation leads to the selective lysosomal clearance of MUT TP53, highlighting a potential therapeutic vulnerability of aggressive tumors expressing this oncoprotein.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":""},"PeriodicalIF":14.3,"publicationDate":"2025-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145304922","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-15DOI: 10.1080/15548627.2025.2572530
Virginia Actis Dato, Kyohei Fujita, Stephan Lange
{"title":"Novel links of ATG4 proteases to cytoskeletal adapters of the obscurin protein family.","authors":"Virginia Actis Dato, Kyohei Fujita, Stephan Lange","doi":"10.1080/15548627.2025.2572530","DOIUrl":"https://doi.org/10.1080/15548627.2025.2572530","url":null,"abstract":"<p><p>The obscurin family, containing the giant protein OBSCN (obscurin, cytoskeletal calmodulin and titin-interacting RhoGEF) and its closely related OBSL1 (obscurin like cytoskeletal adaptor 1) as well as SPEG (striated muscle enriched protein kinase) are a group of intracellular proteins that contain serially linked immunoglobulin (Ig) and fibronectin type III (Fn3) domains, along with signaling modules such as protein kinase domains. Hence, obscurins harbor multi-faceted roles for the architecture and organization of cell- and organelle membrane proteins. Besides mediating cellular signaling and promoting protein homeostasis, obscurin proteins are also proposed to act as versatile cytoskeletal linkers. Due to their close homology, many functions for OBSCN are evolutionary conserved in OBSL1 and SPEG. However, their expression patterns differ widely, with OBSL1 being ubiquitously expressed in all cell types, while OBSCN and SPEG are more restricted to cross-striated muscles. Recent evidence indicates that autophagy-linked peptidases of the ATG4 family interact with the cytoskeletal adapter proteins OBSL1 and OBSCN. Peptidases of the ATG4 family process Atg8-family proteins (e.g. LC3) in their immature state (i.e. as pro-peptides like pro-LC3) or their bioactive lipidated state (i.e. LC3-II) and facilitate their conversion to the delipidated state (i.e. LC3-I). Loss of interaction between ATG4 peptidases and obscurin family proteins affects cellular macroautophagy/autophagy and mitophagy, leading in situations of cellular stress to depletion of ATG4 and accumulation of the lipidated state for Atg8-family proteins (e.g. LC3-II).</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-3"},"PeriodicalIF":14.3,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145304954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The autophagy-recessive tissue hormone DBI/ACBP (diazepam binding inhibitor, acyl-CoA binding protein) contributes to the pathogenesis of osteoarthritis.","authors":"Uxía Nogueira-Recalde, Beatriz Caramés, Isabelle Martins, Guido Kroemer","doi":"10.1080/15548627.2025.2573220","DOIUrl":"https://doi.org/10.1080/15548627.2025.2573220","url":null,"abstract":"<p><p>Osteoarthritis (OA) is the most common form of arthritis and a leading cause of disability in the elderly, characterized by the progressive destruction of cartilage, synovial inflammation, and subchondral bone remodeling. While mechanical stress, metabolic derangements, and systemic inflammation are recognized contributors, accumulating evidence underscores the pivotal role of impaired macroautophagy/autophagy in disease pathogenesis. Autophagy declines with age, depriving chondrocytes and synovial cells of their cytoprotective capacity and rendering them vulnerable to apoptosis, matrix degradation, and inflammatory activation. Recent work has identified DBI/ACBP (diazepam binding inhibitor, acyl-CoA binding protein) as an extracellular hormone that represses autophagy through binding to the GABRG2/GABAARγ2 (gamma-aminobutyric type A receptor subunit gamma2) subunit of the GABA type A receptor. Plasma levels of DBI/ACBP are elevated in metabolic syndrome, obesity, diabetes, and aging, all known OA risk factors, and are upregulated in patients at risk of severe OA. In murine models of experimental OA, genetic deletion or antibody-mediated neutralization of DBI/ACBP mitigates joint inflammation, reduces cartilage destruction, and improves functional outcomes. These findings establish DBI/ACBP as a central pathogenic mediator linking aging-associated autophagy decline to osteoarthritis progression. Targeting DBI/ACBP represents a promising strategy to restore tissue homeostasis and modify the natural course of OA, with direct translational potential.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-3"},"PeriodicalIF":14.3,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145294677","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-14DOI: 10.1080/15548627.2025.2574963
Rong Wang, Yi Sun, Jingsong Zhao, Zhongyan Xu, Qi Wei, Tao Zhang, Yanbing Lin, Geng Guo, Xun Li, Wenxin Huang, Jiang Qian, Shuaishuai Xing, Manli Wang, Min Qi, Huidong Zhang
{"title":"Lnc-<i>HZ14</i> promotes LLPS-mediated SPHK1 aggrephagy degradation to suppress trophoblast cell proliferation in unexplained recurrent miscarriage.","authors":"Rong Wang, Yi Sun, Jingsong Zhao, Zhongyan Xu, Qi Wei, Tao Zhang, Yanbing Lin, Geng Guo, Xun Li, Wenxin Huang, Jiang Qian, Shuaishuai Xing, Manli Wang, Min Qi, Huidong Zhang","doi":"10.1080/15548627.2025.2574963","DOIUrl":"https://doi.org/10.1080/15548627.2025.2574963","url":null,"abstract":"<p><p>High incidence of recurrent miscarriage (RM, recurrent abnormal early embryo loss) largely limits global human reproduction. However, it is unclear how the pathogenesis greatly restricts its effective clinical treatment. In our previous studies, we have identified a group of novel long non-coding RNAs (lncRNAs), which might regulate the occurrence of RM through unknown biological mechanisms. In this study, we confirm that a novel lncRNA, lnc-<i>HZ14</i>, which is highly expressed in unexplained RM vs healthy control (HC) villous tissues, is associated with RM using a new RM case-control group (<i>n</i> = 50). In trophoblast cellular assays, lnc-<i>HZ14</i> suppresses trophoblast cell proliferation by specifically downregulating SPHK1 (sphingosine kinase 1) protein levels. In terms of mechanism, lnc-<i>HZ14</i> upregulates SQSTM1/p62 protein levels, enhances its protein interactions with polyubiquitin-modified SPHK1, promotes the formation of SQSTM1-SPHK1 bodies through liquid-liquid phase separation (LLPS), and accelerates SPHK1 aggrephagy degradation. Meanwhile, lnc-<i>HZ14</i> also promotes autophagy by activating ETV4-mediated transcription of <i>ATG101</i> and <i>PPP1R15A/GADD34</i>. The cellular mechanisms are consistent with those in villous tissues of RM patients and in placental tissues of a mouse miscarriage model, excepting that there is no lnc-<i>HZ14</i> homolog in mouse. As for miscarriage treatment, therapeutic upregulation of SPHK1 by treatment with phorbol 12-myristate 13-acetate (PMA), an SPHK1 agonist recovers mouse placental proliferation and alleviates mouse miscarriage. Collectively, this study shows for the first time the regulatory roles of lnc-<i>HZ14</i>, LLPS, and aggrephagy degradation of SPHK1 in unexplained recurrent miscarriage, uncovering novel pathogenesis and biological mechanisms of unexplained RM and also providing potential targets for treatment against miscarriage.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":""},"PeriodicalIF":14.3,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145288015","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-14DOI: 10.1080/15548627.2025.2568487
Jinghong Yang, Haobin Sun, Keqing Xu, Xiaomei Zhang, Mudan Huang, Guanghui Jin, Yasong Liu, Weizhao Chen, Shunan Lin, Juan Shen, Chuan-Qi Zhong, Yan Xu, Qi Zhang, Wei Liu, Yang Yang, Jingxing Ou
{"title":"Aging-rewired metabolic cues promote autophagy and senescence via DRAM1.","authors":"Jinghong Yang, Haobin Sun, Keqing Xu, Xiaomei Zhang, Mudan Huang, Guanghui Jin, Yasong Liu, Weizhao Chen, Shunan Lin, Juan Shen, Chuan-Qi Zhong, Yan Xu, Qi Zhang, Wei Liu, Yang Yang, Jingxing Ou","doi":"10.1080/15548627.2025.2568487","DOIUrl":"10.1080/15548627.2025.2568487","url":null,"abstract":"<p><p>Being a major contributor to cell senescence and aging, DNA damage activates macroautophagy/autophagy, but how this process is affected by aging-rewired metabolism in normal biological systems remains to be explored. Here in cultured human umbilical cord-derived mesenchymal stem cells (HsMSCs) and the mouse liver that accumulate DNA damage during aging, we found an elevation of DRAM1 (DNA damage regulated autophagy modulator 1) and DRAM1-mediated pro-senescent autophagy (DMPA). Confirming that DRAM1 activated AMPK, we sought DMPA-associated metabolic features and noted substantial enrichment of N-acetylhistamine (N-AcHA) and phosphatidylethanolamine (PE) products in the aging HsMSCs and mouse liver. Elevating DNA damage and senescence, N-AcHA supplements were sufficient to upregulate DRAM1 and DMPA in primary hepatocytes from young mice but not even in pre-senescent HsMSCs, hence reflecting the differential tolerance of these cell models toward cytotoxic metabolic cues. The effects of N-AcHA were further verified in mouse aging and post-hepatectomy liver regeneration models. In contrast, accumulating cellular PE contents via ethanolamine supplements augmented autophagy but not DNA damage and senescence despite tending to induce DRAM1. Combined treatments with N-AcHA and ethanolamine were sufficient to trigger DMPA in HsMSCs. Despite their differential cellular responses toward N-AcHA and ethanolamine supplements, in primary HsMSCs and mouse hepatocytes DMPA did not notably downregulate SQSTM1/p62 proteins, which differed from general macroautophagy and may constitutively support the fusion of SQSTM1-modified cargo-containing autophagosomes with lysosomes. Overall, this study reveals DMPA-promoting metabolic and molecular features. Thus, targeting certain metabolic pathways and DMPA may promote DNA repair and delay senescence/aging.<b>Abbreviations:</b> ATM: ATM serine/threonine kinase; ATG5: autophagy related 5; ACTB: actin beta; BaFA1: bafilomycin A<sub>1</sub>; CDKN1A/p21: cyclin dependent kinase inhibitor 1A; DDR: DNA damage response; DEGs: differentially expressed genes; DRAM1: DNA damage regulated autophagy modulator 1; DMPA: DRAM1-mediated pro-senescent autophagy; DPMPs: differentially presented metabolic products; ETO: etoposide; Eth: ethanolamine; GL: glycerolipids; GP: glycerophospholipids; γ-H2AX: phosphorylated H2A.X variant histone; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HsMSC: human mesenchymal stem cell; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MSA: methanesulfonic acid; N-AcHA: N-acetylhistamine; PE: phosphatidylethanolamine; PHx: partial hepatectomy; PCYT2: phosphate cytidyltransferase 2, ethanolamine; SASP: senescence-associated secretory phenotype; SA-GLB1/β-gal: senescence-associated galactosidase beta 1; SQSTM1/p62: sequestosome 1; TAF: telomere-associated foci; TP53/p53: tumor protein p53.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-23"},"PeriodicalIF":14.3,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145214587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-13DOI: 10.1080/15548627.2025.2572528
Ningning Li, Hongyu Hou, Dan Su, Bojun Yang, Rui Ni, Guoqiang Ma, Shan Sun, Qilian Ma, Qiang Peng, Siqian Chen, Kin Yip Tam, Hongfeng Wang, Zheng Ying
{"title":"PPP2/PP2A-mediated dephosphorylation of LC3B links PINK1-PRKN/Parkin-mediated mitophagy to SCA12 pathogenesis.","authors":"Ningning Li, Hongyu Hou, Dan Su, Bojun Yang, Rui Ni, Guoqiang Ma, Shan Sun, Qilian Ma, Qiang Peng, Siqian Chen, Kin Yip Tam, Hongfeng Wang, Zheng Ying","doi":"10.1080/15548627.2025.2572528","DOIUrl":"10.1080/15548627.2025.2572528","url":null,"abstract":"<p><p>Atg8-family proteins are autophagosome-associated proteins and play important roles in macroautophagy/autophagy, a conserved process for degrading defective or excessive cellular components. Post-translational modifications of mammalian Atg8-family proteins, including phosphorylation, regulate multiple steps in the autophagic process. In this context, several Atg8-family protein-associated kinases have been found to regulate autophagy, yet the phosphatases in the dephosphorylation of Atg8-family proteins remain unknown. Here, we report that the heterotrimeric PPP2/PP2A (protein phosphatase 2) is a novel regulator in modulating LC3B dephosphorylation. Mechanistically, we find that PPP2-mediated LC3B dephosphorylation reduces the interaction between LC3B and the mitophagy receptor OPTN, thereby impeding the mitochondrial recruitment of phagophores during PINK1-PRKN/Parkin-mediated mitophagy. Interestingly, we find that overexpression of the β2 isoform of PPP2R2B (protein phosphatase 2 regulatory subunit Bbeta; PPP2R2Bβ2), which mimics the spinocerebellar ataxia type 12 (SCA12) pathological condition, harms neuronal survival by enhancing PPP2-mediated LC3B dephosphorylation and reducing mitochondrial recruitment of phagophores upon mitochondrial damage. Importantly, pharmacological induction of mitophagy by the small molecule compound deferiprone (DFP) relieves PPP2R2Bβ2-mediated neuronal toxicity. Overall, our results not only uncover a mechanism by which protein dephosphorylation negatively regulates mitophagy but also provide insights into the pathogenesis of PPP2R2Bβ2-mediated SCA12.<b>Abbreviations</b>: AO: antimycin A and oligomycin A; DFP: deferiprone; EGFP: enhanced green fluorescent protein; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; OPTN: optineurin; PINK1: PTEN induced kinase 1; PLA: proximity ligation assay; PPP2: protein phosphatase 2; PPP2CA: protein phosphatase 2 catalytic subunit alpha; PPP2CB: protein phosphatase 2 catalytic subunit beta; PPP2R2Bβ2: protein phosphatase 2 regulatory subunit B beta 2; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; SCA12: spinocerebellar ataxia type 12; WT: wild type.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-13"},"PeriodicalIF":14.3,"publicationDate":"2025-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145254153","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-13DOI: 10.1080/15548627.2025.2572527
Yingying Cong, Fulvio Reggiori
{"title":"Hijacking a cellular highway: non-lipidated LC3 proteins and PCNT (pericentrin) drive influenza a virus uncoating.","authors":"Yingying Cong, Fulvio Reggiori","doi":"10.1080/15548627.2025.2572527","DOIUrl":"10.1080/15548627.2025.2572527","url":null,"abstract":"<p><p>MAP1LC3/LC3 (microtubule associated protein 1 light chain 3) proteins have long been thought to carry out their cellular and organismal functions, including macroautophagy/autophagy, exclusively in their lipidated form, also referred to as Atg8ylation. They are anchored mainly to the phosphatidylethanolamine present in membranes through the action of two ubiquitin-like conjugation systems. Our recent work, however, uncovered a role of non-lipidated LC3s during influenza A virus (IAV) infection. We revealed that LC3s, together with the centrosomal scaffold protein PCNT (pericentrin), form a dynein adaptor complex that facilitates IAV uncoating at late endosomes (LEs). We also showed that co-opting the LC3s-PCNT complex is an alternative strategy to aggresome processing machinery (APM) hijacking via HDAC6, allowing IAV to exploit the force generated by dynein-dependent motors for virion uncoating and genome delivery in the host cytoplasm. Notably, the function of LC3s in IAV uncoating does not require their Atg8ylation or the core autophagy machinery, and PCNT's role is independent from its centrosomal localization. These findings redefine LC3s as multifunctional adaptor proteins and reveal how viruses can co-opt centrosome assembly machinery components for host invasion.<b>Abbreviation</b>: AKAP9/AKAP450- A-kinase anchoring protein 9; APM- aggresome processing machinery; IAV- influenza A virus; LC3s-I- non-lipidated LC3s; Les- late endosomes; MAP1LC3/LC3s-microtubule associated protein 1 light chain 3 proteins; MT-microtubule; NEU- neuraminidase; PCNT-pericentrin; TNPO1-transportin 1; vRNP-viral ribonucleoprotein.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-3"},"PeriodicalIF":14.3,"publicationDate":"2025-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145254193","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-10DOI: 10.1080/15548627.2025.2569965
Hyun Jung Park, JiYeon Kim, Jiwoo Choi, Chongsuk Ryou, Eunji Shin, Jae Young Lee
{"title":"Targeted genome editing of ZKSCAN3 mitigates the neurotoxicity caused by mutant HTT (huntingtin) in a Huntington disease animal model and three-dimensional cell culture of Huntington disease.","authors":"Hyun Jung Park, JiYeon Kim, Jiwoo Choi, Chongsuk Ryou, Eunji Shin, Jae Young Lee","doi":"10.1080/15548627.2025.2569965","DOIUrl":"10.1080/15548627.2025.2569965","url":null,"abstract":"<p><p>Huntington disease (HD) is a neurodegenerative disease caused by the expression of a mutant form of HTT (huntingtin; mHTT), caused by an abnormal expansion of polyglutamine in HTT. In HD, macroautophagy/autophagy dysfunction can cause mHTT accumulation. Moreover, the promotion of autophagy is considered a therapeutic strategy for the treatment of HD. ZKSCAN3 (zinc finger with KRAB And SCAN domains 3) has been identified as a transcriptional repressor of TFEB (transcription factor EB), a master regulator of autophagy and lysosomal functions. In this study, we conducted CRISPR-Cas9-based gene ablation to disrupt ZKSCAN3 in HD animal models and HD patient-induced pluripotent stem cell (iPSC) -derived three-dimensional (3D) spheroids. In animal models of HD, targeted in vivo <i>zkscan3</i> ablation via a single adeno-associated virus (AAV) mediated CRISPR-Cas9 approach resulted in reduced mHTT levels, leading to improvements in both behavioral symptoms and the brain environment. Furthermore, CRISPR-Cas9 mediated ablation of ZKSCAN3 in 3D spheroids from HD patient-derived iPSC resulted in increased autophagy and lysosomal function, along with reduced mHTT accumulation. Specifically, in iPSC-derived neurons from HD patients, ZKSCAN3-depleted neurons demonstrated increased lysosomal function and reduced oxidative stress compared to controls. Additionally, transcriptional analysis of ZKSCAN3-edited neurons revealed an increased expression of genes involved in synaptic function and transporter activity. Taken together, these results suggest that in HD treatment strategies for improving neuronal function and the brain environment, ZKSCAN3 downregulation in neurons by autophagy activation may improve the brain environment through neuronal self-repair.<b>Abbreviations:</b> 2D: two-dimensional; 3D: three-dimensional; 4-HNE: 4-hydroxynonenal; AAV: adeno-associated virus; AD: Alzheimer disease; Aβ: beta-amyloid; DAPI: 4,6-diamidino-2-phenylindole; GFP: green fluorescent protein; HD: Huntington disease; HTT: huntingtin; IXMC: ImageXpress microconfocal high-content imaging system; Indel: insertion or deletion; iPSC: induced pluripotent stem cell; LAMP1: lysosomal-associated membrane protein 1; mHTT: mutant huntingtin; NPCs: neural precursor cells; RBFOX3/NeuN: RNA binding fox-1 homolog 3; PD: Parkinson disease; RNP: ribonucleoprotein; sgRNAs: single guide RNAs; ST: striatum; TFEB: transcription factor EB; TUBB3/Tuj-1: tubulin beta 3 class III; ZKSCAN3: zinc finger with KRAB and SCAN domains 3.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-15"},"PeriodicalIF":14.3,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145214755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutophagyPub Date : 2025-10-10DOI: 10.1080/15548627.2025.2572026
Rui Liu, Tamar Avin-Wittenberg, Qingqiu Gong
{"title":"Design and validation of a synthetic chlorophagy receptor.","authors":"Rui Liu, Tamar Avin-Wittenberg, Qingqiu Gong","doi":"10.1080/15548627.2025.2572026","DOIUrl":"https://doi.org/10.1080/15548627.2025.2572026","url":null,"abstract":"<p><p>Chloroplasts are photosynthetic, protein-rich organelles unique to green plants and algae. In more complex plants, each mesophyll cell may contain up to 100 chloroplasts, each 5 - 10 µm in length, and 50% of plant proteins are stored in chloroplasts as RUBISCO. Plant cells clear parts of or entire chloroplasts by piecemeal autophagy, microautophagy, and other vesicular pathways to maintain homeostasis and to recycle nutrients in developmental and stress conditions. However, canonical, membrane-bound chlorophagy receptors remain unidentified, and whether chlorophagy can be induced to enhance plant fitness remains unclear. In a recent study, we designed and validated a synthetic chlorophagy receptor, LIR-SNT-BFP. This receptor, upon dexamethasone (DEX) induction, recruits ATG8 to chloroplasts and promotes chloroplast microautophagy independent of ATG5 and ATG7, and induces chloroplast fission. Expression of this synthetic receptor helps protect chloroplasts and seedlings from herbicide damage. Moderate induction of this synthetic receptor promotes rosette growth, while excessive induction inhibits growth and reduces chlorophyll levels. There is a clear upper limit to the benefits of inducing autophagy in plants. This synthetic chlorophagy receptor may help identify endogenous receptors and key players in microautophagy, and can be a powerful new tool for studies investigating chloroplast dynamics and nutrient remobilization.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-3"},"PeriodicalIF":14.3,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145276892","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}