首页 > 最新文献

Autophagy最新文献

英文 中文
PINK1-deficiency facilitates mitochondrial iron accumulation and colon tumorigenesis. PINK1 缺失会促进线粒体铁积累和结肠肿瘤发生。
Pub Date : 2024-11-16 DOI: 10.1080/15548627.2024.2425594
Mariella Arcos, Lavanya Goodla, Hyeoncheol Kim, Sharina P Desai, Rui Liu, Kunlun Yin, Zhaoli Liu, David R Martin, Xiang Xue
<p><p>Mitophagy, the process by which cells eliminate damaged mitochondria, is mediated by PINK1 (PTEN induced kinase 1). Our recent research indicates that PINK1 functions as a tumor suppressor in colorectal cancer by regulating cellular metabolism. Interestingly, PINK1 ablation activated the NLRP3 (NLR family pyrin domain containing 3) inflammasome, releasing IL1B (interleukin 1 beta). However, inhibiting the NLRP3-IL1B signaling pathway with an IL1R (interleukin 1 receptor) antagonist or NLRP3 inhibitor did not hinder colon tumor growth after PINK1 loss. To identify druggable targets in PINK1-deficient tumors, ribonucleic acid sequencing analysis was performed on colon tumors from <i>pink1</i> knockout and wild-type mice. Gene Set Enrichment Analysis highlighted the enrichment of iron ion transmembrane transporter activity. Subsequent qualitative polymerase chain reaction and western blot analysis revealed an increase in mitochondrial iron transporters, including mitochondrial calcium uniporter, in PINK1-deficient colon tumor cells and tissues. Live-cell iron staining demonstrated elevated cellular and mitochondrial iron levels in PINK1-deficient cells. Clinically used drugs deferiprone and minocycline reduced mitochondrial iron and superoxide levels, resulting in decreased colon tumor cell growth <i>in vitro</i> and <i>in vivo</i>. Manipulating the mitochondrial iron uptake protein MCU (mitochondrial calcium uniporter) also affected cell and xenograft tumor growth. This study suggests that therapies aimed at reducing mitochondrial iron levels may effectively inhibit colon tumor growth, particularly in patients with low PINK1 expression.<b>Abbreviation</b>: ANOVA: analysis of variance; APC: adenomatous polyposis coli; cAMP: cyclic adenosine monophosphate; CDX2: caudal type homeobox 2; CGAS: cyclic GMP-AMP synthase; CRC: colorectal cancer; DNA: deoxyribonucleic acid; DFP: deferiprone; DMEM: Dulbecco's modified Eagle medium; DSS: dextran sodium sulfate; ERT2-Cre: Cre recombinase fused to a triple mutant form of the human estrogen receptor; EV: empty vector; GLB: glybenclamide/glyburide; H&E: hematoxylin and eosin; ICP-MS: inductively coupled plasma mass spectrometer; IL1B: interleukin 1 beta; kDa: kilodalton; MCU: mitochondrial calcium uniporter; MKI67: marker of proliferation Ki-67; mRNA: messenger ribonucleic acid; MTT: 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide; NLRP3: NLR family pyrin domain containing 3; OE: overexpression; PBS: phosphate-buffered saline; p-CREB: phosphorylated cAMP responsive element binding protein; PINK1: PTEN induced kinase 1; p-PRKAA/AMPK: phosphorylated protein kinase AMP-activated catalytic subunit alpha; qPCR: qualitative polymerase chain reaction; RNA-seq: ribonucleic acid sequencing; ROS: reactive oxygen species; sg: single guide; sh: short hairpin; SLC25A28: solute carrier family 25 member 28; SLC25A37/MFRN: solute carrier family 25 member 37; STING1: stimulator of interferon response cGAMP inte
细胞消除受损线粒体的过程是由 PINK1(PTEN 诱导激酶 1)介导的。我们最近的研究表明,PINK1 通过调节细胞新陈代谢在结直肠癌中发挥肿瘤抑制因子的功能。有趣的是,PINK1 消融会激活 NLRP3(NLR 家族含吡咯啉结构域 3)炎性体,释放 IL1B(白细胞介素 1 beta)。然而,使用IL1R(白细胞介素1受体)拮抗剂或NLRP3抑制剂抑制NLRP3-IL1B信号通路并不能阻止PINK1缺失后结肠肿瘤的生长。为了确定 PINK1 缺失肿瘤中的药物靶点,对粉红 1 基因敲除小鼠和野生型小鼠的结肠肿瘤进行了核糖核酸测序分析。基因组富集分析突显了铁离子跨膜转运体活性的富集。随后的定性聚合酶链反应和 Western 印迹分析显示,在 PINK1 基因缺陷的结肠肿瘤细胞和组织中,线粒体铁转运体(包括线粒体钙离子单转运体)有所增加。活细胞铁染色显示,PINK1 基因缺陷细胞中的细胞和线粒体铁含量升高。临床常用药物去铁酮和米诺环素可降低线粒体铁和超氧化物水平,从而减少结肠肿瘤细胞在体外和体内的生长。操纵线粒体铁摄取蛋白 MCU(线粒体钙单运蛋白)也会影响细胞和异种移植肿瘤的生长。这项研究表明,旨在降低线粒体铁含量的疗法可有效抑制结肠肿瘤的生长,尤其是在 PINK1 表达量较低的患者中。
{"title":"PINK1-deficiency facilitates mitochondrial iron accumulation and colon tumorigenesis.","authors":"Mariella Arcos, Lavanya Goodla, Hyeoncheol Kim, Sharina P Desai, Rui Liu, Kunlun Yin, Zhaoli Liu, David R Martin, Xiang Xue","doi":"10.1080/15548627.2024.2425594","DOIUrl":"10.1080/15548627.2024.2425594","url":null,"abstract":"&lt;p&gt;&lt;p&gt;Mitophagy, the process by which cells eliminate damaged mitochondria, is mediated by PINK1 (PTEN induced kinase 1). Our recent research indicates that PINK1 functions as a tumor suppressor in colorectal cancer by regulating cellular metabolism. Interestingly, PINK1 ablation activated the NLRP3 (NLR family pyrin domain containing 3) inflammasome, releasing IL1B (interleukin 1 beta). However, inhibiting the NLRP3-IL1B signaling pathway with an IL1R (interleukin 1 receptor) antagonist or NLRP3 inhibitor did not hinder colon tumor growth after PINK1 loss. To identify druggable targets in PINK1-deficient tumors, ribonucleic acid sequencing analysis was performed on colon tumors from &lt;i&gt;pink1&lt;/i&gt; knockout and wild-type mice. Gene Set Enrichment Analysis highlighted the enrichment of iron ion transmembrane transporter activity. Subsequent qualitative polymerase chain reaction and western blot analysis revealed an increase in mitochondrial iron transporters, including mitochondrial calcium uniporter, in PINK1-deficient colon tumor cells and tissues. Live-cell iron staining demonstrated elevated cellular and mitochondrial iron levels in PINK1-deficient cells. Clinically used drugs deferiprone and minocycline reduced mitochondrial iron and superoxide levels, resulting in decreased colon tumor cell growth &lt;i&gt;in vitro&lt;/i&gt; and &lt;i&gt;in vivo&lt;/i&gt;. Manipulating the mitochondrial iron uptake protein MCU (mitochondrial calcium uniporter) also affected cell and xenograft tumor growth. This study suggests that therapies aimed at reducing mitochondrial iron levels may effectively inhibit colon tumor growth, particularly in patients with low PINK1 expression.&lt;b&gt;Abbreviation&lt;/b&gt;: ANOVA: analysis of variance; APC: adenomatous polyposis coli; cAMP: cyclic adenosine monophosphate; CDX2: caudal type homeobox 2; CGAS: cyclic GMP-AMP synthase; CRC: colorectal cancer; DNA: deoxyribonucleic acid; DFP: deferiprone; DMEM: Dulbecco's modified Eagle medium; DSS: dextran sodium sulfate; ERT2-Cre: Cre recombinase fused to a triple mutant form of the human estrogen receptor; EV: empty vector; GLB: glybenclamide/glyburide; H&E: hematoxylin and eosin; ICP-MS: inductively coupled plasma mass spectrometer; IL1B: interleukin 1 beta; kDa: kilodalton; MCU: mitochondrial calcium uniporter; MKI67: marker of proliferation Ki-67; mRNA: messenger ribonucleic acid; MTT: 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide; NLRP3: NLR family pyrin domain containing 3; OE: overexpression; PBS: phosphate-buffered saline; p-CREB: phosphorylated cAMP responsive element binding protein; PINK1: PTEN induced kinase 1; p-PRKAA/AMPK: phosphorylated protein kinase AMP-activated catalytic subunit alpha; qPCR: qualitative polymerase chain reaction; RNA-seq: ribonucleic acid sequencing; ROS: reactive oxygen species; sg: single guide; sh: short hairpin; SLC25A28: solute carrier family 25 member 28; SLC25A37/MFRN: solute carrier family 25 member 37; STING1: stimulator of interferon response cGAMP inte","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-17"},"PeriodicalIF":0.0,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142607142","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}
引用次数: 0
Deciphering melanophagy: role of the PTK2-ITCH-MLANA-OPTN cascade on melanophagy in melanocytes. 解密黑色素吞噬:PTK2-ITCH-MLANA-OPTN 级联在黑色素细胞黑色素吞噬中的作用。
Pub Date : 2024-11-12 DOI: 10.1080/15548627.2024.2421695
Na Yeon Park, Doo Sin Jo, Hyun Jun Park, Ji-Eun Bae, Yong Hwan Kim, Joon Bum Kim, Ha Jung Lee, Sung Hyun Kim, Hyunjung Choi, Hyun-Shik Lee, Tamotsu Yoshimori, Dong-Seok Lee, Jin-A Lee, Pansoo Kim, Dong-Hyung Cho

Melanosomes play a pivotal role in skin color and photoprotection. In contrast to the well-elucidated pathway of melanosome biogenesis, the process of melanosome degradation, referred to as melanophagy, is largely unexplored. Previously, we discovered that 3,4,5-trimethoxycinnamate thymol ester (TCTE) effectively inhibits skin pigmentation by activating melanophagy. In this study, we discovered a new regulatory signaling cascade that controls melanophagy in TCTE-treated melanocytes. ITCH (itchy E3 ubiquitin protein ligase) facilitates ubiquitination of the melanosome membrane protein MLANA (melan-A) during TCTE-induced melanophagy. This ubiquitinated MLANA is then recognized by an autophagy receptor protein, OPTN (optineurin). Additionally, a phospho-kinase antibody array revealed that TCTE activates PTK2 (protein tyrosine kinase 2), which phosphorylates ITCH, enhancing the ubiquitination of MLANA. Furthermore, inhibition of either PTK2 or ITCH disrupts the ubiquitination of MLANA and the MLANA-OPTN interaction in TCTE-treated cells. Taken together, our findings highlight the critical role of the PTK2-ITCH-MLANA-OPTN cascade in orchestrating melanophagy progression.Abbreviations: α-MSH: alpha-melanocyte-stimulating hormone; dichlone: 2,3-dichloro-1,4-naphthoquinone; ITCH: itchy E3 ubiquitin protein ligase; MITF: melanocyte inducing transcription factor; MLANA: melan-A; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PINK1: PTEN induced kinase 1; PTK2: protein tyrosine kinase 2; SQSTM1/p62: sequestosome 1; TCTE: 3,4,5-trimethoxycinnamate thymol ester; TPC2: two pore segment channel 2; VDAC1: voltage dependent anion channel 1.

黑色素体在肤色和光保护方面起着关键作用。与已阐明的黑色素小体生物生成途径不同,黑色素小体的降解过程(即黑色素吞噬)在很大程度上尚未被探索。此前,我们发现 3,4,5-三甲氧基肉桂酸胸腺酚酯(TCTE)能通过激活黑色素吞噬作用有效抑制皮肤色素沉着。在这项研究中,我们发现了一种新的调控信号级联,它能控制经 TCTE 处理的黑色素细胞中的黑色素吞噬。在TCTE诱导的黑色素吞噬过程中,ITCH(痒E3泛素蛋白连接酶)促进了黑色素体膜蛋白MLANA(melan-A)的泛素化。泛素化后的 MLANA 会被自噬受体蛋白 OPTN(optineurin)识别。此外,磷酸激酶抗体阵列显示,TCTE 能激活 PTK2(蛋白酪氨酸激酶 2),使 ITCH 磷酸化,从而增强 MLANA 的泛素化。此外,抑制 PTK2 或 ITCH 会破坏 TCTE 处理细胞中 MLANA 的泛素化和 MLANA-OPTN 的相互作用。综上所述,我们的研究结果凸显了 PTK2-ITCH-MLANA-OPTN 级联在协调黑色素吞噬过程中的关键作用。
{"title":"Deciphering melanophagy: role of the PTK2-ITCH-MLANA-OPTN cascade on melanophagy in melanocytes.","authors":"Na Yeon Park, Doo Sin Jo, Hyun Jun Park, Ji-Eun Bae, Yong Hwan Kim, Joon Bum Kim, Ha Jung Lee, Sung Hyun Kim, Hyunjung Choi, Hyun-Shik Lee, Tamotsu Yoshimori, Dong-Seok Lee, Jin-A Lee, Pansoo Kim, Dong-Hyung Cho","doi":"10.1080/15548627.2024.2421695","DOIUrl":"10.1080/15548627.2024.2421695","url":null,"abstract":"<p><p>Melanosomes play a pivotal role in skin color and photoprotection. In contrast to the well-elucidated pathway of melanosome biogenesis, the process of melanosome degradation, referred to as melanophagy, is largely unexplored. Previously, we discovered that 3,4,5-trimethoxycinnamate thymol ester (TCTE) effectively inhibits skin pigmentation by activating melanophagy. In this study, we discovered a new regulatory signaling cascade that controls melanophagy in TCTE-treated melanocytes. ITCH (itchy E3 ubiquitin protein ligase) facilitates ubiquitination of the melanosome membrane protein MLANA (melan-A) during TCTE-induced melanophagy. This ubiquitinated MLANA is then recognized by an autophagy receptor protein, OPTN (optineurin). Additionally, a phospho-kinase antibody array revealed that TCTE activates PTK2 (protein tyrosine kinase 2), which phosphorylates ITCH, enhancing the ubiquitination of MLANA. Furthermore, inhibition of either PTK2 or ITCH disrupts the ubiquitination of MLANA and the MLANA-OPTN interaction in TCTE-treated cells. Taken together, our findings highlight the critical role of the PTK2-ITCH-MLANA-OPTN cascade in orchestrating melanophagy progression.<b>Abbreviations</b>: α-MSH: alpha-melanocyte-stimulating hormone; dichlone: 2,3-dichloro-1,4-naphthoquinone; ITCH: itchy E3 ubiquitin protein ligase; MITF: melanocyte inducing transcription factor; MLANA: melan-A; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PINK1: PTEN induced kinase 1; PTK2: protein tyrosine kinase 2; SQSTM1/p62: sequestosome 1; TCTE: 3,4,5-trimethoxycinnamate thymol ester; TPC2: two pore segment channel 2; VDAC1: voltage dependent anion channel 1.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-10"},"PeriodicalIF":0.0,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549513","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}
引用次数: 0
HSP90 N-terminal inhibition promotes mitochondria-derived vesicles related metastasis by reducing TFEB transcription via decreased HSP90AA1-HCFC1 interaction in liver cancer. 在肝癌中,HSP90 N-端抑制通过减少HSP90AA1与HCFC1的相互作用,降低TFEB转录,从而促进线粒体衍生囊泡的相关转移。
Pub Date : 2024-11-11 DOI: 10.1080/15548627.2024.2421703
Lixia Liu, Zhenming Zheng, Yaling Huang, Hairou Su, Guibing Wu, Zihao Deng, Yan Li, Guantai Xie, Jieyou Li, Fei Zou, Xuemei Chen

Cancer cells compensate with increasing mitochondria-derived vesicles (MDVs) to maintain mitochondrial homeostasis, when canonical MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta)-mediated mitophagy is lacking. MDVs promote the transport of mitochondrial components into extracellular vesicles (EVs) and induce tumor metastasis. Although HSP90 (heat shock protein 90) chaperones hundreds of client proteins and its inhibitors suppress tumors, HSP90 inhibitors-related chemotherapy is associated with unexpected metastasis. Herein, we find that HSP90 inhibitor causes mitochondrial damage but stimulates the low LC3-induced MDVs and the release of MDVs-derived EVs. However, why LC3 decreases and what is the transcriptional regulatory mechanism of MDVs formation under HSP90 inhibition remain unknown. Because TFEB (transcription factor EB) is the most important mitophagy transcription factor, and the HSP90 client HCFC1 (host cell factor C1) regulates TFEB transcription, there should be a hidden connection between TFEB, HCFC1 and HSP90 in MDVs formation. Our results support the idea that HSP90 N-terminal inhibition reduces TFEB transcription via decreased HSP90AA1-HCFC1 interaction, which prevents HCFC1 from binding to the TFEB proximal promoter region. Decreased TFEB transcription and consequently reduced LC3, ultimately promoted MDVs formation. Blocking MDVs formation with the microtubule inhibitor nocodazole (NOC) activates the HCFC1-TFEB-LC3 axis, weakens HSP90 inhibitors-induced MDVs and the release of MDVs-derived EVs, inhibits the growth of tumor cell spheres and primary liver tumors, and reduces the extravasation of cancer cells to secondary metastatic sites. Taken together, these data suggest that combination therapy should be used to reduce the metastatic risk of low TFEB-triggered-MDVs formation caused by HSP90 inhibitors.Abbreviation: ACIs: ATP-competitive inhibitors; BaFA1: bafilomycin A1; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; ChIP: chromatin immunoprecipitation; CHX: cycloheximide; CTD: C-terminal domain; EVs: extracellular vesicles; HCFC1: host cell factor C1; HSP90: heat shock protein 90; ILVs: intralumenal vesicles; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MD: middle domain; MDVs: mitochondria-derived vesicles; MQC: mitochondrial quality control; ΔΨm: mitochondrial membrane potential; MVBs: multivesicular bodies; NB: novobiocin; TEM: transmission electron microscopy; TFEB: transcription factor EB; TFs: transcription factors. NOC: nocodazole; NTD: N-terminal nucleotide binding domain; OCR: oxygen consumption rate; RFP: red fluorescent protein; ROS: reactive oxygen species; STA9090: Ganetespib; VPS35: VPS35 retromer complex component.

当缺乏典型的 MAP1LC3B/LC3B(微管相关蛋白 1 轻链 3 beta)介导的有丝分裂时,癌细胞会通过增加线粒体衍生囊泡(MDVs)来维持线粒体的平衡。MDV 促进线粒体成分向细胞外囊泡 (EV) 的运输,并诱发肿瘤转移。虽然 HSP90(热休克蛋白 90)能合成数百种客户蛋白,其抑制剂也能抑制肿瘤,但与 HSP90 抑制剂相关的化疗却会导致意想不到的转移。在这里,我们发现 HSP90 抑制剂会导致线粒体损伤,但会刺激低 LC3 诱导的 MDVs 和 MDVs 衍生 EVs 的释放。然而,在 HSP90 抑制作用下,LC3 为什么会减少,MDVs 形成的转录调控机制是什么,这些仍然是未知数。由于TFEB(转录因子EB)是最重要的有丝分裂吞噬转录因子,而HSP90的客户HCFC1(宿主细胞因子C1)调控TFEB的转录,因此在MDVs形成过程中,TFEB、HCFC1和HSP90之间应该存在隐性联系。我们的研究结果支持这样一种观点,即 HSP90 N 端抑制通过减少 HSP90AA1 与 HCFC1 的相互作用来减少 TFEB 的转录,从而阻止 HCFC1 与 TFEB 近端启动子区域结合。TFEB 转录的减少以及 LC3 的减少最终促进了 MDVs 的形成。用微管抑制剂nocodazole(NOC)阻断MDVs的形成可激活HCFC1-TFEB-LC3轴,削弱HSP90抑制剂诱导的MDVs和MDVs衍生EVs的释放,抑制肿瘤细胞球和原发性肝肿瘤的生长,并减少癌细胞向继发性转移部位的外渗。综上所述,这些数据表明,应采用联合疗法来降低 HSP90 抑制剂导致的低 TFEB 触发 MDVs 形成的转移风险。
{"title":"HSP90 N-terminal inhibition promotes mitochondria-derived vesicles related metastasis by reducing TFEB transcription via decreased HSP90AA1-HCFC1 interaction in liver cancer.","authors":"Lixia Liu, Zhenming Zheng, Yaling Huang, Hairou Su, Guibing Wu, Zihao Deng, Yan Li, Guantai Xie, Jieyou Li, Fei Zou, Xuemei Chen","doi":"10.1080/15548627.2024.2421703","DOIUrl":"10.1080/15548627.2024.2421703","url":null,"abstract":"<p><p>Cancer cells compensate with increasing mitochondria-derived vesicles (MDVs) to maintain mitochondrial homeostasis, when canonical MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta)-mediated mitophagy is lacking. MDVs promote the transport of mitochondrial components into extracellular vesicles (EVs) and induce tumor metastasis. Although HSP90 (heat shock protein 90) chaperones hundreds of client proteins and its inhibitors suppress tumors, HSP90 inhibitors-related chemotherapy is associated with unexpected metastasis. Herein, we find that HSP90 inhibitor causes mitochondrial damage but stimulates the low LC3-induced MDVs and the release of MDVs-derived EVs. However, why LC3 decreases and what is the transcriptional regulatory mechanism of MDVs formation under HSP90 inhibition remain unknown. Because TFEB (transcription factor EB) is the most important mitophagy transcription factor, and the HSP90 client HCFC1 (host cell factor C1) regulates <i>TFEB</i> transcription, there should be a hidden connection between TFEB, HCFC1 and HSP90 in MDVs formation. Our results support the idea that HSP90 N-terminal inhibition reduces <i>TFEB</i> transcription via decreased HSP90AA1-HCFC1 interaction, which prevents HCFC1 from binding to the <i>TFEB</i> proximal promoter region. Decreased <i>TFEB</i> transcription and consequently reduced LC3, ultimately promoted MDVs formation. Blocking MDVs formation with the microtubule inhibitor nocodazole (NOC) activates the HCFC1-<i>TFEB</i>-LC3 axis, weakens HSP90 inhibitors-induced MDVs and the release of MDVs-derived EVs, inhibits the growth of tumor cell spheres and primary liver tumors, and reduces the extravasation of cancer cells to secondary metastatic sites. Taken together, these data suggest that combination therapy should be used to reduce the metastatic risk of low <i>TFEB</i>-triggered-MDVs formation caused by HSP90 inhibitors.<b>Abbreviation</b>: ACIs: ATP-competitive inhibitors; BaFA1: bafilomycin A1; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; ChIP: chromatin immunoprecipitation; CHX: cycloheximide; CTD: C-terminal domain; EVs: extracellular vesicles; HCFC1: host cell factor C1; HSP90: heat shock protein 90; ILVs: intralumenal vesicles; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MD: middle domain; MDVs: mitochondria-derived vesicles; MQC: mitochondrial quality control; ΔΨm: mitochondrial membrane potential; MVBs: multivesicular bodies; NB: novobiocin; TEM: transmission electron microscopy; TFEB: transcription factor EB; TFs: transcription factors. NOC: nocodazole; NTD: N-terminal nucleotide binding domain; OCR: oxygen consumption rate; RFP: red fluorescent protein; ROS: reactive oxygen species; STA9090: Ganetespib; VPS35: VPS35 retromer complex component.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-25"},"PeriodicalIF":0.0,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142514559","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}
引用次数: 0
Atractylenolide I inhibits angiogenesis and reverses sunitinib resistance in clear cell renal cell carcinoma through ATP6V0D2-mediated autophagic degradation of EPAS1/HIF2α. 白术内酯I通过ATP6V0D2介导的EPAS1/HIF2α自噬降解抑制血管生成并逆转透明细胞肾细胞癌的舒尼替尼耐药性。
Pub Date : 2024-11-04 DOI: 10.1080/15548627.2024.2421699
Qinyu Li, Kai Zeng, Qian Chen, Chenglin Han, Xi Wang, Beining Li, Jianping Miao, Bolong Zheng, Jihong Liu, Xianglin Yuan, Bo Liu

Clear cell renal cell carcinoma (ccRCC) is tightly associated with VHL (von Hippel-Lindau tumor suppressor) mutation and dysregulated angiogenesis. Accumulating evidence indicates that antiangiogenic treatment abolishing tumor angiogenesis can achieve longer disease-free survival in patients with ccRCC. Atractylenolide I (ATL-I) is one of the main active compounds in Atractylodes macrocephala root extract and exhibits various pharmacological effects, including anti-inflammatory and antitumor effects. In this study, we revealed the potent antitumor activity of ATL-I in ccRCC. ATL-I exhibited robust antiangiogenic capacity by inhibiting EPAS1/HIF2α-mediated VEGFA production in VHL-deficient ccRCC, and it promoted autophagic degradation of EPAS1 by upregulating the ATPase subunit ATP6V0D2 (ATPase H+ transporting V0 subunit d2) to increase lysosomal function and facilitated fusion between autophagosomes and lysosomes. Mechanistically, ATP6V0D2 directly bound to RAB7 and VPS41 and promoted the RAB7-HOPS interaction, facilitating SNARE complex assembly and autophagosome-lysosome fusion. Moreover, ATP6V0D2 promoted autolysosome degradation by increasing the acidification and activity of lysosomes during the later stages of macroautophagy/autophagy. Additionally, we found that ATL-I could decrease the level of EPAS1, which was upregulated in sunitinib-resistant cells, thus reversing sunitinib resistance. Collectively, our findings demonstrate that ATL-I is a robust antiangiogenic and antitumor lead compound with potential clinical application for ccRCC therapy.Abbreviations: ATL-I: atractylenolide I; ATP6V0D2: ATPase H+ transporting V0 subunit d2; CAM: chick chorioallantoic membrane; ccRCC: clear cell renal cell carcinoma; CTSB: cathepsin B; CTSD: cathepsin D; GO: Gene Ontology; HIF-1: HIF1A-ARNT heterodimer; HOPS: homotypic fusion and protein sorting; KDR/VEGFR: kinase insert domain receptor; KEGG: Kyoto Encyclopedia of Genes and Genomes; RCC: renal cell carcinoma; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; TCGA: The Cancer Genome Atlas; TEM: transmission electron microscopy; TKI: tyrosine kinase inhibitor; V-ATPase: vacuolar-type H±translocating ATPase; VEGF: vascular endothelial growth factor; VHL: von Hippel-Lindau tumor suppressor.

透明细胞肾细胞癌(ccRCC)与VHL(von Hippel-Lindau肿瘤抑制因子)突变和血管生成失调密切相关。越来越多的证据表明,消除肿瘤血管生成的抗血管生成治疗可以延长ccRCC患者的无病生存期。白术内酯 I(ATL-I)是白术根提取物中的主要活性化合物之一,具有多种药理作用,包括抗炎和抗肿瘤作用。本研究揭示了 ATL-I 在 ccRCC 中的强效抗肿瘤活性。ATL-I通过抑制EPAS1/HIF2α介导的VEGFA生成,在VHL缺陷的ccRCC中表现出强大的抗血管生成能力,并通过上调ATP酶亚基ATP6V0D2(ATP酶H+转运V0亚基d2)促进EPAS1的自噬降解,从而增强溶酶体功能,促进自噬体和溶酶体之间的融合。从机制上讲,ATP6V0D2直接与RAB7和VPS41结合,促进了RAB7-HOPS的相互作用,有利于SNARE复合物的组装和自噬体与溶酶体的融合。此外,在大自噬/自噬的后期阶段,ATP6V0D2通过提高溶酶体的酸化和活性促进自溶酶体降解。此外,我们还发现 ATL-I 能降低舒尼替尼耐药细胞中上调的 EPAS1 水平,从而逆转舒尼替尼耐药。总之,我们的研究结果表明,ATL-I 是一种强效的抗血管生成和抗肿瘤先导化合物,具有临床应用于 ccRCC 治疗的潜力。
{"title":"Atractylenolide I inhibits angiogenesis and reverses sunitinib resistance in clear cell renal cell carcinoma through ATP6V0D2-mediated autophagic degradation of EPAS1/HIF2α.","authors":"Qinyu Li, Kai Zeng, Qian Chen, Chenglin Han, Xi Wang, Beining Li, Jianping Miao, Bolong Zheng, Jihong Liu, Xianglin Yuan, Bo Liu","doi":"10.1080/15548627.2024.2421699","DOIUrl":"10.1080/15548627.2024.2421699","url":null,"abstract":"<p><p>Clear cell renal cell carcinoma (ccRCC) is tightly associated with <i>VHL</i> (von Hippel-Lindau tumor suppressor) mutation and dysregulated angiogenesis. Accumulating evidence indicates that antiangiogenic treatment abolishing tumor angiogenesis can achieve longer disease-free survival in patients with ccRCC. Atractylenolide I (ATL-I) is one of the main active compounds in <i>Atractylodes macrocephala</i> root extract and exhibits various pharmacological effects, including anti-inflammatory and antitumor effects. In this study, we revealed the potent antitumor activity of ATL-I in ccRCC. ATL-I exhibited robust antiangiogenic capacity by inhibiting EPAS1/HIF2α-mediated VEGFA production in VHL-deficient ccRCC, and it promoted autophagic degradation of EPAS1 by upregulating the ATPase subunit ATP6V0D2 (ATPase H+ transporting V0 subunit d2) to increase lysosomal function and facilitated fusion between autophagosomes and lysosomes. Mechanistically, ATP6V0D2 directly bound to RAB7 and VPS41 and promoted the RAB7-HOPS interaction, facilitating SNARE complex assembly and autophagosome-lysosome fusion. Moreover, ATP6V0D2 promoted autolysosome degradation by increasing the acidification and activity of lysosomes during the later stages of macroautophagy/autophagy. Additionally, we found that ATL-I could decrease the level of EPAS1, which was upregulated in sunitinib-resistant cells, thus reversing sunitinib resistance. Collectively, our findings demonstrate that ATL-I is a robust antiangiogenic and antitumor lead compound with potential clinical application for ccRCC therapy.<b>Abbreviations</b>: ATL-I: atractylenolide I; ATP6V0D2: ATPase H+ transporting V0 subunit d2; CAM: chick chorioallantoic membrane; ccRCC: clear cell renal cell carcinoma; CTSB: cathepsin B; CTSD: cathepsin D; GO: Gene Ontology; HIF-1: HIF1A-ARNT heterodimer; HOPS: homotypic fusion and protein sorting; KDR/VEGFR: kinase insert domain receptor; KEGG: Kyoto Encyclopedia of Genes and Genomes; RCC: renal cell carcinoma; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; TCGA: The Cancer Genome Atlas; TEM: transmission electron microscopy; TKI: tyrosine kinase inhibitor; V-ATPase: vacuolar-type H±translocating ATPase; VEGF: vascular endothelial growth factor; VHL: von Hippel-Lindau tumor suppressor.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"1-20"},"PeriodicalIF":0.0,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549512","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}
引用次数: 0
Recycling the recyclers: lysophagy emerges as a new pharmacological target for retinal degeneration. 回收者的回收:溶酶体成为治疗视网膜变性的新药理靶点。
Pub Date : 2024-11-01 Epub Date: 2024-08-17 DOI: 10.1080/15548627.2024.2391726
Juan Ignacio Jiménez-Loygorri, Patricia Boya

Dysregulated macroautophagy/autophagy is one of the hallmarks of aging and has also been linked to higher incidence of several age-associated diseases such as age-related macular degeneration (AMD). The main cell type affected in AMD is the retinal pigment epithelium (RPE), and this disease can lead to central vision loss. Despite affecting around 8.7% of the population between 45-85 years, its etiopathogenesis remains unknown. In our recent manuscript using the pharmacological sodium iodate (SI) model of AMD we identified severe lysosomal membrane permeabilization (LMP) in the RPE, that leads to autophagy flux blockage and proteostasis defects. Treatment with the natural compound urolithin A (UA) reduces RPE cell death and alleviates vision loss, concurrent with full autophagy restoration. While UA was initially described as a specific mitophagy inducer, we now show that it is also able to promote SQSTM1/p62-dependent lysophagy in the context of lysosomal damage and LMP. Genetic downregulation of SQSTM1/p62 fully abolishes the effect of UA on lysophagy while mitophagy stimulation remains unaffected. In summary, these findings highlight the wide range of pathways modulated by UA and its potential implementation in the management of AMD and other diseases involving lysosomal damage.

大自噬/自噬失调是衰老的标志之一,也与几种老年相关疾病(如老年性黄斑变性(AMD))的高发病率有关。受老年黄斑变性影响的主要细胞类型是视网膜色素上皮(RPE),这种疾病会导致中心视力丧失。尽管在 45-85 岁的人群中,约有 8.7% 的人患有这种疾病,但其发病机理仍然不明。在我们最近的手稿中,我们使用药理碘酸钠(SI)模型来研究老年性黄斑病变,发现 RPE 中存在严重的溶酶体膜通透性(LMP),这会导致自噬通路受阻和蛋白稳态缺陷。使用天然化合物尿石素 A(UA)治疗可减少 RPE 细胞死亡,缓解视力丧失,同时自噬功能也得到了完全恢复。虽然 UA 最初被描述为一种特异性有丝分裂诱导剂,但我们现在发现它还能在溶酶体损伤和 LMP 的情况下促进 SQSTM1/p62 依赖性溶酶体吞噬。基因下调 SQSTM1/p62 能完全消除 UA 对溶酶吞噬的影响,而对有丝分裂的刺激则不受影响。总之,这些发现凸显了尿崩症调节的广泛途径及其在治疗老年性黄斑变性和其他涉及溶酶体损伤的疾病中的应用潜力。
{"title":"Recycling the recyclers: lysophagy emerges as a new pharmacological target for retinal degeneration.","authors":"Juan Ignacio Jiménez-Loygorri, Patricia Boya","doi":"10.1080/15548627.2024.2391726","DOIUrl":"10.1080/15548627.2024.2391726","url":null,"abstract":"<p><p>Dysregulated macroautophagy/autophagy is one of the hallmarks of aging and has also been linked to higher incidence of several age-associated diseases such as age-related macular degeneration (AMD). The main cell type affected in AMD is the retinal pigment epithelium (RPE), and this disease can lead to central vision loss. Despite affecting around 8.7% of the population between 45-85 years, its etiopathogenesis remains unknown. In our recent manuscript using the pharmacological sodium iodate (SI) model of AMD we identified severe lysosomal membrane permeabilization (LMP) in the RPE, that leads to autophagy flux blockage and proteostasis defects. Treatment with the natural compound urolithin A (UA) reduces RPE cell death and alleviates vision loss, concurrent with full autophagy restoration. While UA was initially described as a specific mitophagy inducer, we now show that it is also able to promote SQSTM1/p62-dependent lysophagy in the context of lysosomal damage and LMP. Genetic downregulation of SQSTM1/p62 fully abolishes the effect of UA on lysophagy while mitophagy stimulation remains unaffected. In summary, these findings highlight the wide range of pathways modulated by UA and its potential implementation in the management of AMD and other diseases involving lysosomal damage.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"2589-2590"},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572288/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141918324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dysregulation of pancreatic β-cell autophagy and the risk of type 2 diabetes. 胰腺β细胞自噬失调与2型糖尿病的风险
Pub Date : 2024-11-01 Epub Date: 2024-06-18 DOI: 10.1080/15548627.2024.2367356
Hayder M Al-Kuraishy, Majid S Jabir, Ali I Al-Gareeb, Daniel J Klionsky, Ali K Albuhadily

Macroautophagy/autophagy is an essential degradation process that removes abnormal cellular components, maintains homeostasis within cells, and provides nutrition during starvation. Activated autophagy enhances cell survival during stressful conditions, although overactivation of autophagy triggers induction of autophagic cell death. Therefore, early-onset autophagy promotes cell survival whereas late-onset autophagy provokes programmed cell death, which can prevent disease progression. Moreover, autophagy regulates pancreatic β-cell functions by different mechanisms, although the precise role of autophagy in type 2 diabetes (T2D) is not completely understood. Consequently, this mini-review discusses the protective and harmful roles of autophagy in the pancreatic β cell and in the pathophysiology of T2D.

自噬是一种重要的降解过程,可清除异常细胞成分,维持细胞内的平衡,并在饥饿时提供营养。激活自噬可提高细胞在压力条件下的存活率,但过度激活自噬会诱导自噬细胞死亡。因此,早期发生的自噬可促进细胞存活,而晚期发生的自噬会导致细胞程序性死亡,从而阻止不同疾病的进展。此外,自噬通过不同的机制调节胰腺β细胞的功能,但自噬在2型糖尿病(T2D)中的确切作用尚不完全清楚。因此,这篇微型综述讨论了自噬在胰腺β细胞和T2D病理生理学中的保护和有害作用。
{"title":"Dysregulation of pancreatic β-cell autophagy and the risk of type 2 diabetes.","authors":"Hayder M Al-Kuraishy, Majid S Jabir, Ali I Al-Gareeb, Daniel J Klionsky, Ali K Albuhadily","doi":"10.1080/15548627.2024.2367356","DOIUrl":"10.1080/15548627.2024.2367356","url":null,"abstract":"<p><p>Macroautophagy/autophagy is an essential degradation process that removes abnormal cellular components, maintains homeostasis within cells, and provides nutrition during starvation. Activated autophagy enhances cell survival during stressful conditions, although overactivation of autophagy triggers induction of autophagic cell death. Therefore, early-onset autophagy promotes cell survival whereas late-onset autophagy provokes programmed cell death, which can prevent disease progression. Moreover, autophagy regulates pancreatic β-cell functions by different mechanisms, although the precise role of autophagy in type 2 diabetes (T2D) is not completely understood. Consequently, this mini-review discusses the protective and harmful roles of autophagy in the pancreatic β cell and in the pathophysiology of T2D.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"2361-2372"},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572262/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141319257","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The DNA damage response and autophagy during cancer development: an antagonistic pleiotropy entanglement. 癌症发展过程中的 DNA 损伤反应和自噬:一种拮抗的多义性纠缠。
Pub Date : 2024-11-01 Epub Date: 2024-06-10 DOI: 10.1080/15548627.2024.2362121
Vassilis G Gorgoulis, Konstantinos Evangelou, Daniel J Klionsky

The DNA damage response (DDR) pathway is a cardinal cellular stress response mechanism that during cancer development follows an antagonistic pleiotropy mode of action. Given that DDR activation is an energy demanding process, interplay with macroautophagy/autophagy, a stress response and energy providing mechanism, is likely to take place. While molecular connections between both mechanisms have been reported, an open question regards whether autophagy activation follows solely or is entangled with DDR in a similar antagonistic pleiotropy pattern during cancer development. Combing evidence on the spatiotemporal relationship of DDR and autophagy in the entire spectrum of carcinogenesis from our previous studies, we discuss these issues in the current addendum.Abbreviation: AMPK: AMP-dependent protein kinase; DDR: DNA damage response.

DNA 损伤应答(DDR)途径是一种重要的细胞应激反应机制,在癌症发展过程中遵循一种拮抗性多效应作用模式。鉴于 DDR 激活是一个需要能量的过程,因此很可能会与作为应激反应和能量提供机制的大自噬/自噬发生相互作用。虽然这两种机制之间的分子联系已有报道,但一个悬而未决的问题是,在癌症的发展过程中,自噬的激活是完全跟随 DDR 还是与 DDR 以类似的拮抗褶皱模式纠缠在一起。我们在本增编中讨论了这些问题,并结合以往研究中有关 DDR 和自噬在整个癌变过程中的时空关系的证据。
{"title":"The DNA damage response and autophagy during cancer development: an antagonistic pleiotropy entanglement.","authors":"Vassilis G Gorgoulis, Konstantinos Evangelou, Daniel J Klionsky","doi":"10.1080/15548627.2024.2362121","DOIUrl":"10.1080/15548627.2024.2362121","url":null,"abstract":"<p><p>The DNA damage response (DDR) pathway is a cardinal cellular stress response mechanism that during cancer development follows an antagonistic pleiotropy mode of action. Given that DDR activation is an energy demanding process, interplay with macroautophagy/autophagy, a stress response and energy providing mechanism, is likely to take place. While molecular connections between both mechanisms have been reported, an open question regards whether autophagy activation follows solely or is entangled with DDR in a similar antagonistic pleiotropy pattern during cancer development. Combing evidence on the spatiotemporal relationship of DDR and autophagy in the entire spectrum of carcinogenesis from our previous studies, we discuss these issues in the current addendum.<b>Abbreviation</b>: AMPK: AMP-dependent protein kinase; DDR: DNA damage response.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"2571-2573"},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572190/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141201582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lassa virus Z protein hijacks the autophagy machinery for efficient transportation by interrupting CCT2-mediated cytoskeleton network formation. 拉沙病毒 Z 蛋白通过干扰 CCT2 介导的细胞骨架网络形成,劫持自噬机制以实现高效运输。
Pub Date : 2024-11-01 Epub Date: 2024-07-20 DOI: 10.1080/15548627.2024.2379099
Yueming Yuan, An Fang, Mai Zhang, Ming Zhou, Zhen F Fu, Ling Zhao

The Lassa virus (LASV) is a widely recognized virulent pathogen that frequently results in lethal viral hemorrhagic fever (VHF). Earlier research has indicated that macroautophagy/autophagy plays a role in LASV replication, but, the precise mechanism is unknown. In this present study, we show that LASV matrix protein (LASV-Z) is essential for blocking intracellular autophagic flux. LASV-Z hinders actin and tubulin folding by interacting with CCT2, a component of the chaperonin-containing T-complexes (TRiC). When the cytoskeleton is disrupted, lysosomal enzyme transit is hampered. In addition, cytoskeleton disruption inhibits the merge of autophagosomes with lysosomes, resulting in autophagosome accumulation that promotes the budding of LASV virus-like particles (VLPs). Inhibition of LASV-Z-induced autophagosome accumulation blocks the LASV VLP budding process. Furthermore, it is found that glutamine at position 29 and tyrosine at position 48 on LASV-Z are important in interacting with CCT2. When these two sites are mutated, LASV-mut interacts with CCT2 less efficiently and can no longer inhibit the autophagic flux. These findings demonstrate a novel strategy for LASV-Z to hijack the host autophagy machinery to accomplish effective transportation.Abbreviation: 3-MA: 3-methyladenine; ATG5: autophagy related 5; ATG7: autophagy related 7; Baf-A1: bafilomycin A1; CCT2: chaperonin containing TCP1 subunit 2; co-IP: co-immunoprecipitation; CTSD: cathepsin D; DAPI: 4',6-diamidino-2'-phenylindole; DMSO: dimethyl sulfoxide; EGFR: epidermal growth factor receptor; GFP: green fluorescent protein; hpi: hours post-infection; hpt: hours post-transfection; LAMP1: lysosomal-associated membrane protein 1; LASV: lassa virus; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mCherry: red fluorescent protein; PM: plasma membrane; SQSTM1/p62: sequestosome 1; STX6: syntaxin 6; VLP: virus-like particle; TEM: transmission electron microscopy; TRiC: chaperonin-containing T-complex; WB: western blotting; μm: micrometer; μM: micromole.

拉沙病毒(LASV)是一种公认的致病性病原体,经常导致致命的病毒性出血热(VHF)。早先的研究表明,大自噬/自噬在 LASV 复制过程中发挥作用,但其确切机制尚不清楚。在本研究中,我们发现 LASV 基质蛋白(LASV-Z)对阻断细胞内自噬通量至关重要。LASV-Z 通过与含伴侣素 T-复合体(TRiC)的一个成分 CCT2 相互作用,阻碍肌动蛋白和微管蛋白折叠。当细胞骨架被破坏时,溶酶体酶的转运就会受到阻碍。此外,细胞骨架破坏会抑制自噬体与溶酶体的合并,导致自噬体堆积,从而促进 LASV 病毒样颗粒(VLP)的出芽。抑制 LASV-Z 诱导的自噬体积累会阻止 LASV VLP 的出芽过程。此外,研究还发现,LASV-Z 上第 29 位的谷氨酰胺和第 48 位的酪氨酸在与 CCT2 的相互作用中非常重要。当这两个位点发生突变时,LASV-mut 与 CCT2 的相互作用效率降低,不能再抑制自噬通量。这些发现表明,LASV-Z 采用了一种新策略来劫持宿主的自噬机制,从而完成有效的运输。
{"title":"Lassa virus Z protein hijacks the autophagy machinery for efficient transportation by interrupting CCT2-mediated cytoskeleton network formation.","authors":"Yueming Yuan, An Fang, Mai Zhang, Ming Zhou, Zhen F Fu, Ling Zhao","doi":"10.1080/15548627.2024.2379099","DOIUrl":"10.1080/15548627.2024.2379099","url":null,"abstract":"<p><p>The Lassa virus (LASV) is a widely recognized virulent pathogen that frequently results in lethal viral hemorrhagic fever (VHF). Earlier research has indicated that macroautophagy/autophagy plays a role in LASV replication, but, the precise mechanism is unknown. In this present study, we show that LASV matrix protein (LASV-Z) is essential for blocking intracellular autophagic flux. LASV-Z hinders actin and tubulin folding by interacting with CCT2, a component of the chaperonin-containing T-complexes (TRiC). When the cytoskeleton is disrupted, lysosomal enzyme transit is hampered. In addition, cytoskeleton disruption inhibits the merge of autophagosomes with lysosomes, resulting in autophagosome accumulation that promotes the budding of LASV virus-like particles (VLPs). Inhibition of LASV-Z-induced autophagosome accumulation blocks the LASV VLP budding process. Furthermore, it is found that glutamine at position 29 and tyrosine at position 48 on LASV-Z are important in interacting with CCT2. When these two sites are mutated, LASV-mut interacts with CCT2 less efficiently and can no longer inhibit the autophagic flux. These findings demonstrate a novel strategy for LASV-Z to hijack the host autophagy machinery to accomplish effective transportation.<b>Abbreviation:</b> 3-MA: 3-methyladenine; ATG5: autophagy related 5; ATG7: autophagy related 7; Baf-A1: bafilomycin A<sub>1</sub>; CCT2: chaperonin containing TCP1 subunit 2; co-IP: co-immunoprecipitation; CTSD: cathepsin D; DAPI: 4',6-diamidino-2'-phenylindole; DMSO: dimethyl sulfoxide; EGFR: epidermal growth factor receptor; GFP: green fluorescent protein; hpi: hours post-infection; hpt: hours post-transfection; LAMP1: lysosomal-associated membrane protein 1; LASV: lassa virus; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mCherry: red fluorescent protein; PM: plasma membrane; SQSTM1/p62: sequestosome 1; STX6: syntaxin 6; VLP: virus-like particle; TEM: transmission electron microscopy; TRiC: chaperonin-containing T-complex; WB: western blotting; μm: micrometer; μM: micromole.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"2511-2528"},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572193/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141617785","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Signal-Sustained Imaging of Mitophagy with an Enzyme-Activatable Metabolic Lipid Labeling Probe. 利用酶促代谢脂质标记探针对有丝分裂进行信号持续成像。
Pub Date : 2024-11-01 Epub Date: 2024-06-19 DOI: 10.1080/15548627.2024.2367192
Xiaoxue Zou, Shixiong Wen, Lichun Xu, Lei Gao, Xunxiang Wang, Xiao Hu, Jiahuai Han, Shoufa Han

Imaging of mitophagy is of significance as aberrant mitophagy is engaged in multiple diseases. Mitophagy has been imaged with synthetic or biotic pH sensors by reporting pH acidification en route delivery into lysosomes. To circumvent uncertainty of acidity-dependent signals, we herein report an enzyme-activatable probe covalently attached on mitochondrial inner membrane (ECAM) for signal-persist mitophagy imaging. ECAM is operated via ΔΨm-driven accumulation of Mito-proGreen in mitochondria and covalent linking of the trapped probe with azidophospholipids metabolically incorporated into the mitochondrial inner membrane. Upon mitophagy, ECAM is delivered into lysosomes and hydrolyzed by LNPEP/leucyl aminopeptidase, yielding turn-on green fluorescence that is immune to lysosomal acidity changes and stably retained in fixed cells. With ECAM, phorbol-12-myristate-13-acetate (PMA) was identified as a highly potent inducer of mitophagy. Overcoming signal susceptibility of pH probes and liability of ΔΨm probes to dissipation from stressed mitochondria, ECAM offers an attractive tool to study mitophagy and mitophagy-inducing therapeutic agents.Abbreviations: Baf-A1, bafilomycin A1; CCCP, carbonyl cyanide m-chlorophenylhydrazone; DBCO, dibenzocyclooctyne; ECAM, enzyme-activated probe covalently attached on mitochondrial inner membrane; GFP, green fluorescent protein; LAMP2, lysosomal associated membrane protein 2; LNPEP/LAP, leucyl and cystinyl aminopeptidase; PMA, phorbol-12-myristate-13-acetate; ΔΨm, mitochondrial transmembrane potential; RFP, red fluorescent protein; TPP, triphenylphosphonium.

有丝分裂成像具有重要意义,因为有丝分裂异常与多种疾病有关。有丝分裂是通过合成或生物 pH 传感器报告进入溶酶体途中的 pH 酸化来成像的。为了避免酸度依赖性信号的不确定性,我们在此报告了一种共价连接在线粒体内膜(ECAM)上的酶激活探针,用于信号持久的有丝分裂成像。ECAM是通过ΔΨm驱动线粒体中Mito-proGreen的积累和被捕获探针与线粒体内膜中代谢结合的叠氮磷脂的共价连接来运行的。在有丝分裂过程中,ECAM 被送入溶酶体并被 LNPEP/白氨酰基氨肽酶水解,从而产生绿色荧光,这种荧光不受溶酶体酸度变化的影响,并能稳定地保留在固定的细胞中。通过 ECAM,发现光稳定剂-12-肉豆蔻酸-13-乙酸酯(PMA)是一种高效的有丝分裂诱导剂。ECAM克服了pH探针的信号易感性和ΔΨm探针从受压线粒体中消散的特性,为研究有丝分裂和有丝分裂诱导治疗剂提供了一种极具吸引力的工具。
{"title":"Signal-Sustained Imaging of Mitophagy with an Enzyme-Activatable Metabolic Lipid Labeling Probe.","authors":"Xiaoxue Zou, Shixiong Wen, Lichun Xu, Lei Gao, Xunxiang Wang, Xiao Hu, Jiahuai Han, Shoufa Han","doi":"10.1080/15548627.2024.2367192","DOIUrl":"10.1080/15548627.2024.2367192","url":null,"abstract":"<p><p>Imaging of mitophagy is of significance as aberrant mitophagy is engaged in multiple diseases. Mitophagy has been imaged with synthetic or biotic pH sensors by reporting pH acidification en route delivery into lysosomes. To circumvent uncertainty of acidity-dependent signals, we herein report an <u>e</u>nzyme-activatable probe <u>c</u>ovalently <u>a</u>ttached on <u>m</u>itochondrial inner membrane (ECAM) for signal-persist mitophagy imaging. ECAM is operated via ΔΨm-driven accumulation of Mito-proGreen in mitochondria and covalent linking of the trapped probe with azidophospholipids metabolically incorporated into the mitochondrial inner membrane. Upon mitophagy, ECAM is delivered into lysosomes and hydrolyzed by LNPEP/leucyl aminopeptidase, yielding turn-on green fluorescence that is immune to lysosomal acidity changes and stably retained in fixed cells. With ECAM, phorbol-12-myristate-13-acetate (PMA) was identified as a highly potent inducer of mitophagy. Overcoming signal susceptibility of pH probes and liability of ΔΨm probes to dissipation from stressed mitochondria, ECAM offers an attractive tool to study mitophagy and mitophagy-inducing therapeutic agents.<b>Abbreviations</b>: Baf-A1, bafilomycin A<sub>1</sub>; CCCP, carbonyl cyanide <i>m</i>-chlorophenylhydrazone; DBCO, dibenzocyclooctyne; ECAM, enzyme-activated probe covalently attached on mitochondrial inner membrane; GFP, green fluorescent protein; LAMP2, lysosomal associated membrane protein 2; LNPEP/LAP, leucyl and cystinyl aminopeptidase; PMA, phorbol-12-myristate-13-acetate; ΔΨm, mitochondrial transmembrane potential; RFP, red fluorescent protein; TPP, triphenylphosphonium.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"2556-2570"},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572071/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141319259","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The autophagy protein RUBCNL/PACER represses RIPK1 kinase-dependent apoptosis and necroptosis. 自噬蛋白RUBCNL/PACER抑制RIPK1激酶依赖的细胞凋亡和坏死。
Pub Date : 2024-11-01 Epub Date: 2024-07-03 DOI: 10.1080/15548627.2024.2367923
Diego Rojas-Rivera, Sebastián Beltrán, Francisco Muñoz-Carvajal, Pablo Ahumada-Montalva, Lorena Abarzúa, Laura Gomez, Fernanda Hernandez, Cristian A Bergmann, Luis Labrador, Melissa Calegaro-Nassif, Mathieu J M Bertrand, Patricio A Manque, Ute Woehlbier

Mesenchymal stem cells (MSCs) are used in cell therapy; nonetheless, their application is limited by their poor survival after transplantation in a proinflammatory microenvironment. Macroautophagy/autophagy activation in MSCs constitutes a stress adaptation pathway, promoting cellular homeostasis. Our proteomics data indicate that RUBCNL/PACER (RUN and cysteine rich domain containing beclin 1 interacting protein like), a positive regulator of autophagy, is also involved in cell death. Hence, we screened MSC survival upon various cell death stimuli under loss or gain of function of RUBCNL. MSCs were protected from TNF (tumor necrosis factor)-induced regulated cell death when RUBCNL was expressed. TNF promotes inflammation by inducing RIPK1 kinase-dependent apoptosis or necroptosis. We determine that MSCs succumb to RIPK1 kinase-dependent apoptosis upon TNF sensing and necroptosis when caspases are inactivated. We show that RUBCNL is a negative regulator of both RIPK1-dependent apoptosis and necroptosis. Furthermore, RUBCNL mutants that lose the ability to regulate autophagy, retain their function in negatively regulating cell death. We also found that RUBCNL forms a complex with RIPK1, which disassembles in response to TNF. In line with this finding, RUBCNL expression limits assembly of RIPK1-TNFRSF1A/TNFR1 complex I, suggesting that complex formation between RUBCNL and RIPK1 represses TNF signaling. These results provide new insights into the crosstalk between the RIPK1-mediated cell death and autophagy machineries and suggest that RUBCNL, due to its functional duality in autophagy and apoptosis/necroptosis, could be targeted to improve the therapeutic efficacy of MSCs. Abbreviations: BAF: bafilomycin A1; CASP3: caspase 3; Caspases: cysteine-aspartic proteases; cCASP3: cleaved CASP3; CQ: chloroquine; CHX: cycloheximide; cPARP: cleaved poly (ADP-ribose) polymerase; DEPs: differential expressed proteins; ETO: etoposide; MEF: mouse embryonic fibroblast; MLKL: mixed lineage kinase domain-like; MSC: mesenchymal stem cell; MTORC1: mechanistic target of rapamycin kinase complex 1; Nec1s: necrostatin 1s; NFKB/NF-kB: nuclear factor of kappa light polypeptide gene enhancer in B cells; PLA: proximity ligation assay; RCD: regulated cell death; RIPK1: receptor (TNFRSF)-interacting serine-threonine kinase 1; RIPK3: receptor-interacting serine-threonine kinase 3; RUBCNL/PACER: RUN and cysteine rich domain containing beclin 1 interacting protein like; siCtrl: small interfering RNA nonsense; siRNA: small interfering RNA; TdT: terminal deoxynucleotidyl transferase; Tm: tunicamycin; TNF: tumor necrosis factor; TNFRSF1A/TNFR1: tumor necrosis factor receptor superfamily, member 1a.

间充质干细胞(MSCs)被用于细胞治疗;然而,由于它们在促炎症微环境中移植后存活率低,其应用受到了限制。间充质干细胞中的大自噬/自噬激活是一种应激适应途径,可促进细胞稳态。我们的蛋白质组学数据表明,RUBCNL/PACER(RUN and cysteine rich domain containing beclin 1 interacting protein like)是自噬的正调控因子,也参与细胞死亡。因此,我们筛选了在 RUBCNL 功能缺失或增益的情况下,间充质干细胞在各种细胞死亡刺激下的存活率。当 RUBCNL 表达时,间充质干细胞在 TNF(肿瘤坏死因子)诱导的调节性细胞死亡中受到保护。TNF 通过诱导依赖于 RIPK1 激酶的细胞凋亡或坏死来促进炎症。我们确定间充质干细胞在TNF作用下会发生依赖于RIPK1激酶的凋亡,而在caspase失活时会发生坏死。我们发现 RUBCNL 是 RIPK1 依赖性凋亡和坏死的负调控因子。此外,失去自噬调节能力的 RUBCNL 突变体保留了其负向调节细胞死亡的功能。我们还发现,RUBCNL 与 RIPK1 形成了一个复合物,该复合物在 TNF 作用下会解体。与这一发现相一致的是,RUBCNL 的表达限制了 RIPK1-TNFRSF1A/TNFR1 复合物 I 的组装,这表明 RUBCNL 和 RIPK1 之间形成的复合物抑制了 TNF 信号转导。这些结果为研究 RIPK1 介导的细胞死亡和自噬机制之间的相互影响提供了新的视角,并表明 RUBCNL 由于其在自噬和细胞凋亡/坏死中的双重功能,可以作为改善间充质干细胞疗效的靶点。
{"title":"The autophagy protein RUBCNL/PACER represses RIPK1 kinase-dependent apoptosis and necroptosis.","authors":"Diego Rojas-Rivera, Sebastián Beltrán, Francisco Muñoz-Carvajal, Pablo Ahumada-Montalva, Lorena Abarzúa, Laura Gomez, Fernanda Hernandez, Cristian A Bergmann, Luis Labrador, Melissa Calegaro-Nassif, Mathieu J M Bertrand, Patricio A Manque, Ute Woehlbier","doi":"10.1080/15548627.2024.2367923","DOIUrl":"10.1080/15548627.2024.2367923","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) are used in cell therapy; nonetheless, their application is limited by their poor survival after transplantation in a proinflammatory microenvironment. Macroautophagy/autophagy activation in MSCs constitutes a stress adaptation pathway, promoting cellular homeostasis. Our proteomics data indicate that RUBCNL/PACER (RUN and cysteine rich domain containing beclin 1 interacting protein like), a positive regulator of autophagy, is also involved in cell death. Hence, we screened MSC survival upon various cell death stimuli under loss or gain of function of RUBCNL. MSCs were protected from TNF (tumor necrosis factor)-induced regulated cell death when RUBCNL was expressed. TNF promotes inflammation by inducing RIPK1 kinase-dependent apoptosis or necroptosis. We determine that MSCs succumb to RIPK1 kinase-dependent apoptosis upon TNF sensing and necroptosis when caspases are inactivated. We show that RUBCNL is a negative regulator of both RIPK1-dependent apoptosis and necroptosis. Furthermore, RUBCNL mutants that lose the ability to regulate autophagy, retain their function in negatively regulating cell death. We also found that RUBCNL forms a complex with RIPK1, which disassembles in response to TNF. In line with this finding, RUBCNL expression limits assembly of RIPK1-TNFRSF1A/TNFR1 complex I, suggesting that complex formation between RUBCNL and RIPK1 represses TNF signaling. These results provide new insights into the crosstalk between the RIPK1-mediated cell death and autophagy machineries and suggest that RUBCNL, due to its functional duality in autophagy and apoptosis/necroptosis, could be targeted to improve the therapeutic efficacy of MSCs. <b>Abbreviations</b>: BAF: bafilomycin A<sub>1</sub>; CASP3: caspase 3; Caspases: cysteine-aspartic proteases; cCASP3: cleaved CASP3; CQ: chloroquine; CHX: cycloheximide; cPARP: cleaved poly (ADP-ribose) polymerase; DEPs: differential expressed proteins; ETO: etoposide; MEF: mouse embryonic fibroblast; MLKL: mixed lineage kinase domain-like; MSC: mesenchymal stem cell; MTORC1: mechanistic target of rapamycin kinase complex 1; Nec1s: necrostatin 1s; NFKB/NF-kB: nuclear factor of kappa light polypeptide gene enhancer in B cells; PLA: proximity ligation assay; RCD: regulated cell death; RIPK1: receptor (TNFRSF)-interacting serine-threonine kinase 1; RIPK3: receptor-interacting serine-threonine kinase 3; RUBCNL/PACER: RUN and cysteine rich domain containing beclin 1 interacting protein like; si<i>Ctrl</i>: small interfering RNA nonsense; siRNA: small interfering RNA; TdT: terminal deoxynucleotidyl transferase; Tm: tunicamycin; TNF: tumor necrosis factor; TNFRSF1A/TNFR1: tumor necrosis factor receptor superfamily, member 1a.</p>","PeriodicalId":93893,"journal":{"name":"Autophagy","volume":" ","pages":"2444-2459"},"PeriodicalIF":0.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572279/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141319260","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Autophagy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1