1
|
Zeng L, Zhang X, Huang Z, Song S, Li M, Wang T, Sun A, Ge J. Ubiquitin proteasome system in cardiac fibrosis. J Adv Res 2024:S2090-1232(24)00562-9. [PMID: 39653114 DOI: 10.1016/j.jare.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Cardiac fibrosis, including reactive fibrosis and replacement fibrosis, is a common pathological process in most cardiovascular diseases. The ubiquitin proteasome system (UPS) plays an important role in the development of fibrosis by mediating the degradation and synthesis of proteins involved in transforming growth factor-β (TGF-β)-dependent and TGF-β-independent fibrous pathways. AIM OF REVIEW This review aims to provide an overview of ubiquitinated and deubiquitinated molecules that participating in cardiac fibrosis, with the ultimate purpose to identify promising targets for therapeutic strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW The UPS primarily impacts cardiac fibrosis through modulation of the TGF-β signaling pathway targeting key molecules involved, including the TGF-β receptors, Smad2/3/4 complexes, and inhibitory Smad7, thereby influencing fibrotic processes. In addition to its effect on TGF-β signaling, UPS also regulates pro-fibrotic pathways independent of TGF-β, including p53, AKT1-p38, and JNK1/2. Understanding these pathways is critical due to their involvement in diverse fibrotic mechanisms. The interplay between ubiquitination and deubiquitination of crucial pathways and molecules is pivotal in cardiac fibrosis and represents a promising area for identifying novel therapeutic targets. Different types of cardiac fibrosis involve distinct fibrotic pathways, leading to differential effects of ubiquitin ligases (E3 ligases) and deubiquitinating enzymes (DUBs) across various cardiac fibrotic diseases. Insights into UPS-mediated regulation of cardiac fibrosis provide potential anti-fibrotic therapeutic strategies, emphasizing the importance of targeting UPS components specific to the heart for effective therapy against cardiac fibrosis.
Collapse
Affiliation(s)
- Linqi Zeng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaokai Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zihang Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Mohan Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Tongyao Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Edgar K, Simone M, Nina E, Marc S, Farhad S, Jachimowicz RD, Volkmar G, Melanie T. Starvation-induced metabolic rewiring affects mTORC1 composition in vivo. Sci Rep 2024; 14:28296. [PMID: 39550382 PMCID: PMC11569187 DOI: 10.1038/s41598-024-78873-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
Lysosomes play a crucial role in metabolic adaptation to starvation, but detailed in vivo studies are scarce. Therefore, we investigated the changes of the proteome of liver lysosomes in mice starved short-term for 6h or long-term for 24h. We verified starvation-induced catabolism by weight loss, ketone body production, drop in blood glucose and an increase of 3-methylhistidine. Deactivation of mTORC1 in vivo after short-term starvation causes a depletion of mTORC1 and the associated Ragulator complex in hepatic lysosomes, resulting in diminished phosphorylation of mTORC1 target proteins. While mTORC1 lysosomal protein levels and activity in liver were restored after long-term starvation, the lysosomal levels of Ragulator remained constantly reduced. To determine whether this mTORC1 activity pattern may be organ-specific, we further investigated the key metabolic organs muscle and brain. mTORC1 inactivation, but not re-activation, occurred in muscle after a starvation of 12 h or longer. In brain, mTORC1 activity remained unchanged during starvation. As mTORC1 deactivation is known to induce autophagy, we further investigated the more than 150 non-lysosomal proteins enriched in the lysosomal fraction upon starvation. Proteasomal, cytosolic and peroxisomal proteins dominated after short-term starvation, while after long-term starvation, mainly proteasomal and mitochondrial proteins accumulated, indicating ordered autophagic protein degradation.
Collapse
Affiliation(s)
- Kaade Edgar
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Mausbach Simone
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Erps Nina
- Max-Planck Institute for Biology of Ageing, Joseph Stelzmann Str. 9B, 50931, Cologne, Germany
| | - Sylvester Marc
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
- Core Facility Analytical Proteomics, Medical Faculty , Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Shakeri Farhad
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Ron D Jachimowicz
- Max-Planck Institute for Biology of Ageing, Joseph Stelzmann Str. 9B, 50931, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Gieselmann Volkmar
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany
| | - Thelen Melanie
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, 53115, Bonn, Germany.
- Max-Planck Institute for Biology of Ageing, Joseph Stelzmann Str. 9B, 50931, Cologne, Germany.
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany.
| |
Collapse
|
3
|
Ma W, Lu Y, Jin X, Lin N, Zhang L, Song Y. Targeting selective autophagy and beyond: From underlying mechanisms to potential therapies. J Adv Res 2024; 65:297-327. [PMID: 38750694 PMCID: PMC11518956 DOI: 10.1016/j.jare.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Autophagy is an evolutionarily conserved turnover process for intracellular substances in eukaryotes, relying on lysosomal (in animals) or vacuolar (in yeast and plants) mechanisms. In the past two decades, emerging evidence suggests that, under specific conditions, autophagy can target particular macromolecules or organelles for degradation, a process termed selective autophagy. Recently, accumulating studies have demonstrated that the abnormality of selective autophagy is closely associated with the occurrence and progression of many human diseases, including neurodegenerative diseases, cancers, metabolic diseases, and cardiovascular diseases. AIM OF REVIEW This review aims at systematically and comprehensively introducing selective autophagy and its role in various diseases, while unravelling the molecular mechanisms of selective autophagy. By providing a theoretical basis for the development of related small-molecule drugs as well as treating related human diseases, this review seeks to contribute to the understanding of selective autophagy and its therapeutic potential. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we systematically introduce and dissect the major categories of selective autophagy that have been discovered. We also focus on recent advances in understanding the molecular mechanisms underlying both classical and non-classical selective autophagy. Moreover, the current situation of small-molecule drugs targeting different types of selective autophagy is further summarized, providing valuable insights into the discovery of more candidate small-molecule drugs targeting selective autophagy in the future. On the other hand, we also reveal clinically relevant implementations that are potentially related to selective autophagy, such as predictive approaches and treatments tailored to individual patients.
Collapse
Affiliation(s)
- Wei Ma
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xin Jin
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Na Lin
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yaowen Song
- Department of Breast Surgery, Department of Ultrasound, Department of Hematology and Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
4
|
Guo B, Shi X, Jiang Q, Pan Y, Yang Y, Liu Y, Chen S, Zhu W, Ren L, Liang R, Chen X, Xu H, Wei L, Lin Y, Wang J, Qiu C, Zhou H, Rao L, Wang L, Chen R, Chen S. Targeting Immunoproteasome in Polarized Macrophages Ameliorates Experimental Emphysema Via Activating NRF1/2-P62 Axis and Suppressing IRF4 Transcription. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405318. [PMID: 39356034 PMCID: PMC11600198 DOI: 10.1002/advs.202405318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/22/2024] [Indexed: 10/03/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) stands as the prevailing chronic airway ailment, characterized by chronic bronchitis and emphysema. Current medications fall short in treatment of these diseases, underscoring the urgent need for effective therapy. Prior research indicated immunoproteasome inhibition alleviated various inflammatory diseases by modulating immune cell functions. However, its therapeutic potential in COPD remains largely unexplored. Here, an elevated expression of immunoproteasome subunits LMP2 and LMP7 in the macrophages isolated from mouse with LPS/Elastase-induced emphysema and polarized macrophages in vitro is observed. Subsequently, intranasal administration of the immunoproteasome-specific inhibitor ONX-0914 significantly mitigated COPD-associated airway inflammation and improved lung function in mice by suppressing macrophage polarization. Additionally, ONX-0914 capsulated in PLGA nanoparticles exhibited more pronounced therapeutic effect on COPD than naked ONX-0914 by targeting immunoproteasome in polarized macrophages. Mechanistically, ONX-0914 activated autophagy and endoplasmic reticulum (ER) stress are not attribute to the ONX-0914 mediated suppression of macrophage polarization. Intriguingly, ONX-0914 inhibited M1 polarization through the nuclear factor erythroid 2-related factor-1 (NRF1) and NRF2-P62 axis, while the suppression of M2 polarization is regulated by inhibiting the transcription of interferon regulatory factor 4 (IRF4). In summary, the findings suggest that targeting immunoproteasome in macrophages holds promise as a therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Bingxin Guo
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Xing Shi
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Qiong Jiang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Yuanwei Pan
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Yuqiong Yang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory DiseaseFirst Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Yuanyuan Liu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Post‐doctoral Scientific Research Station of Basic Medicine, The Second Clinical Medical CollegeJinan UniversityGuangzhou510632China
| | - Shuyu Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Wenjiao Zhu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Laibin Ren
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Ruifang Liang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Post‐doctoral Scientific Research Station of Basic Medicine, The Second Clinical Medical CollegeJinan UniversityGuangzhou510632China
| | - Xue Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Haizhao Xu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Laiyou Wei
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Yongjian Lin
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Jinyong Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Chen Qiu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Haibo Zhou
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Lang Rao
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Lingwei Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Shanze Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| |
Collapse
|
5
|
Rondón-Ortiz AN, Zhang L, Ash PEA, Basu A, Puri S, van der Spek SJF, Wang Z, Dorrian L, Emili A, Wolozin B. Proximity labeling reveals dynamic changes in the SQSTM1 protein network. J Biol Chem 2024; 300:107621. [PMID: 39098523 PMCID: PMC11401034 DOI: 10.1016/j.jbc.2024.107621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/30/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024] Open
Abstract
Sequestosome1 (SQSTM1) is an autophagy receptor that mediates the degradation of intracellular cargo, including protein aggregates, through multiple protein interactions. These interactions form the SQSTM1 protein network, and these interactions are mediated by SQSTM1 functional interaction domains, which include LIR, PB1, UBA, and KIR. Technological advances in cell biology continue to expand our knowledge of the SQSTM1 protein network and the relationship between the actions of the SQSTM1 protein network in cellular physiology and disease states. Here we apply proximity profile labeling to investigate the SQSTM1 protein interaction network by fusing TurboID with the human protein SQSTM1 (TurboID::SQSTM1). This chimeric protein displayed well-established SQSTM1 features including production of SQSTM1 intracellular bodies, binding to known SQSTM1 interacting partners, and capture of novel SQSTM1 protein interactors. Strikingly, aggregated tau protein altered the protein interaction network of SQSTM1 to include many stress-associated proteins. We demonstrate the importance of the PB1 and/or UBA domains for binding network members, including the K18 domain of tau. Overall, our work reveals the dynamic landscape of the SQSTM1 protein network and offers a resource to study SQSTM1 function in cellular physiology and disease state.
Collapse
Affiliation(s)
- Alejandro N Rondón-Ortiz
- Department of Biology, Boston University, Boston, Massachusetts, USA; Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | - Lushuang Zhang
- Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | - Peter E A Ash
- Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | - Avik Basu
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University, Boston, Massachusetts, USA; Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, Oregon, USA
| | - Sambhavi Puri
- Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | | | - Zihan Wang
- Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | - Luke Dorrian
- Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University, Boston, Massachusetts, USA; Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, Oregon, USA.
| | - Benjamin Wolozin
- Departments of Anatomy & Neurobiology, Boston University, Boston, Massachusetts, USA; Center for Systems Neuroscience, Boston University, Boston, Massachusetts, USA; Center for Neurophotonics, Boston University, Boston, Massachusetts, USA; Department of Neurology, Boston University, Boston, Massachusetts, USA; Department of Pharmacology, Physiology and Biophysics, Boston University, Boston, Massachusetts, USA.
| |
Collapse
|
6
|
Nirmala FS, Lee H, Kim YI, Hahm JH, Seo HD, Kim M, Jung CH, Ahn J. Exercise-induced signaling activation by Chrysanthemum zawadskii and its active compound, linarin, ameliorates age-related sarcopenia through Sestrin 1 regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155695. [PMID: 38728922 DOI: 10.1016/j.phymed.2024.155695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Exercise is an effective strategy to prevent sarcopenia, but high physical inactivity in the elderly requires alternative therapeutic approaches. Exercise mimetics are therapeutic compounds that simulate the beneficial effects of exercise on skeletal muscles. However, the toxicity and adverse effects of exercise mimetics raise serious concerns. PURPOSE We aimed to search novel plant-based alternatives to activate exercise induced-signaling. METHODS We used open databases and luciferase assays to identify plant-derived alternatives to activate exercise-induced signaling and compared its efficacy to mild intensity continuous training (MICT) in aged C57BL/6 mice. The nineteen-month-old mice were either fed an experimental diet supplemented with the isolated alternative or subjected to MICT for up to 21 mo of age. RESULTS Our analysis revealed that Chrysanthemum zawadskii Herbich var latillobum (Maxim.) Kitamura (CZH), a medicinal plant rich in linarin, is a novel activator of peroxisome proliferator-activated receptor δ (PPARδ) and estrogen-related receptor γ (ERRγ), key regulators of exercise-induced positive effects on muscles. CZH supplementation ameliorated the loss of muscle function and mass, and increased PPARδ and ERRγ expression in mouse muscles. CZH also improved mitochondrial functions and proteostasis in aged mice, similar to MICT. Furthermore, CZH and linarin induced the activation of Sestrin 1, a key mediator of exercise benefits, in muscle. Silencing Sestrin 1 negated the increase in myogenesis and mitochondrial respiration by CZH and linarin in primary myoblasts from old mice. CONCLUSION Our findings suggest the potential of CZH as a novel plant-derived alternative to activate exercise-induced signaling for preventing sarcopenia in sedentary older adults. This could offer a safer therapeutic option for sarcopenia treatment.
Collapse
Affiliation(s)
- Farida S Nirmala
- Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea; Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea
| | - Hyunjung Lee
- Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea
| | - Young-In Kim
- Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea
| | - Jeong-Hoon Hahm
- Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea
| | - Hyo-Deok Seo
- Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea
| | - Minjung Kim
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, South Korea
| | - Chang Hwa Jung
- Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea; Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea
| | - Jiyun Ahn
- Department of Food Biotechnology, University of Science and Technology, Daejeon, South Korea; Research Group of Aging and Metabolism, Korea Food Research Institute, Wanju-gun, South Korea.
| |
Collapse
|
7
|
Ortiz ANR, Zhang L, Ash PE, Basu A, Puri S, van der Spek SJ, Wang Z, Dorrian L, Emili A, Wolozin B. Proximity labeling reveals dynamic changes in the SQSTM1 protein network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.12.571324. [PMID: 38168279 PMCID: PMC10760047 DOI: 10.1101/2023.12.12.571324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Sequestosome1 (SQSTM1) is an autophagy receptor that mediates degradation of intracellular cargo, including protein aggregates, through multiple protein interactions. These interactions form the SQSTM1 protein network, and these interactions are mediated by SQSTM1 functional interaction domains, which include LIR, PB1, UBA and KIR. Technological advances in cell biology continue to expand our knowledge of the SQSTM1 protein network and of the relationship of the actions of the SQSTM1 protein network in cellular physiology and disease states. Here we apply proximity profile labeling to investigate the SQSTM1 protein interaction network by fusing TurboID with the human protein SQSTM1 (TurboID::SQSTM1). This chimeric protein displayed well-established SQSTM1 features including production of SQSTM1 intracellular bodies, binding to known SQSTM1 interacting partners, and capture of novel SQSTM1 protein interactors. Strikingly, aggregated tau protein altered the protein interaction network of SQSTM1 to include many stress-associated proteins. We demonstrate the importance of the PB1 and/or UBA domains for binding network members, including the K18 domain of tau. Overall, our work reveals the dynamic landscape of the SQSTM1 protein network and offers a resource to study SQSTM1 function in cellular physiology and disease state.
Collapse
Affiliation(s)
- Alejandro N. Rondón Ortiz
- Department of Biology, Boston University, Boston, MA 02215, USA
- Center for Network Systems Biology, Boston University, Boston, MA 02215, USA
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
| | - Lushuang Zhang
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
| | - Peter E.A. Ash
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
| | - Avik Basu
- Center for Network Systems Biology, Boston University, Boston, MA 02215, USA
- Department of Biochemistry, Boston University, Boston, MA 02115, USA
- Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, OR 97239, USA
| | - Sambhavi Puri
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
| | | | - Zihan Wang
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
| | - Luke Dorrian
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, MA 02215, USA
- Department of Biochemistry, Boston University, Boston, MA 02115, USA
- Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, OR 97239, USA
| | - Benjamin Wolozin
- Departments of Anatomy & Neurobiology, Boston University, Boston, MA 02215, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02115, USA
- Center for Neurophotonics, Boston University, Boston, MA 02115, USA
- Department of Neurology, Boston University, Boston, MA 02115, USA
- Department of Pharmacology, Physiology and Biophysics
| |
Collapse
|
8
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Li G, Chang B, Zhao Y, Wang H, Zhang Y, Zhao M, Zhang L, Song W, Zhang Y. Nano implant surface triggers autophagy through membrane curvature distortion to regulate the osteogenic differentiation. Biomed Mater 2024; 19:035043. [PMID: 38657629 DOI: 10.1088/1748-605x/ad42eb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/24/2024] [Indexed: 04/26/2024]
Abstract
Anodized titania nanotubes have been considered as an effective coating for bone implants due to their ability to induce osteogenesis, whereas the osteogenic mechanism is not fully understood. Our previous study has revealed the potential role of autophagy in osteogenic regulation of nanotubular surface, whereas how the autophagy is activated remains unknown. In this study, we focused on the cell membrane curvature-sensing protein Bif-1 and its effect on the regulation of autophagy. Both autophagosomes formation and autophagic flux were enhanced on the nanotubular surface, as indicated by LC3-II accumulation and p62 degradation. In the meanwhile, the Bif-1 was significantly upregulated, which contributed to autophagy activation and osteogenic differentiation through Beclin-1/PIK3C3 signaling pathway. In conclusion, these findings have bridged the gap between extracellular physical nanotopography and intracellular autophagy activation, which may provide a deeper insight into the signaling transition from mechanical to biological across the cell membrane.
Collapse
Affiliation(s)
- Guangwen Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
- The Affiliated Stomatological Hospital, Southwest Medical University& Institute of Stomatology, Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Bei Chang
- The PLA Rocket Force Characteristic Medical Center, Beijing 100000, People's Republic of China
| | - Yuqi Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
| | - Haochen Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
| | - Yan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
| | - Meiqi Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
| | - Li Zhang
- Department of Prosthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Wen Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
| | - Yumei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xian 710032, People's Republic of China
| |
Collapse
|
10
|
Chakraborty S, Nandi P, Mishra J, Niharika, Roy A, Manna S, Baral T, Mishra P, Mishra PK, Patra SK. Molecular mechanisms in regulation of autophagy and apoptosis in view of epigenetic regulation of genes and involvement of liquid-liquid phase separation. Cancer Lett 2024; 587:216779. [PMID: 38458592 DOI: 10.1016/j.canlet.2024.216779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
Cellular physiology is critically regulated by multiple signaling nexuses, among which cell death mechanisms play crucial roles in controlling the homeostatic landscape at the tissue level within an organism. Apoptosis, also known as programmed cell death, can be induced by external and internal stimuli directing the cells to commit suicide in unfavourable conditions. In contrast, stress conditions like nutrient deprivation, infection and hypoxia trigger autophagy, which is lysosome-mediated processing of damaged cellular organelle for recycling of the degraded products, including amino acids. Apparently, apoptosis and autophagy both are catabolic and tumor-suppressive pathways; apoptosis is essential during development and cancer cell death, while autophagy promotes cell survival under stress. Moreover, autophagy plays dual role during cancer development and progression by facilitating the survival of cancer cells under stressed conditions and inducing death in extreme adversity. Despite having two different molecular mechanisms, both apoptosis and autophagy are interconnected by several crosslinking intermediates. Epigenetic modifications, such as DNA methylation, post-translational modification of histone tails, and miRNA play a pivotal role in regulating genes involved in both autophagy and apoptosis. Both autophagic and apoptotic genes can undergo various epigenetic modifications and promote or inhibit these processes under normal and cancerous conditions. Epigenetic modifiers are uniquely important in controlling the signaling pathways regulating autophagy and apoptosis. Therefore, these epigenetic modifiers of both autophagic and apoptotic genes can act as novel therapeutic targets against cancers. Additionally, liquid-liquid phase separation (LLPS) also modulates the aggregation of misfolded proteins and provokes autophagy in the cytosolic environment. This review deals with the molecular mechanisms of both autophagy and apoptosis including crosstalk between them; emphasizing epigenetic regulation, involvement of LLPS therein, and possible therapeutic approaches against cancers.
Collapse
Affiliation(s)
- Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Prahallad Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bypass Road, Bhauri, Bhopal, 462 030, MP, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India.
| |
Collapse
|
11
|
Li L, Fu S, Wang J, Lu J, Tao Y, Zhao L, Fu B, Lu L, Xiang C, Sun X, Liu S, Wang D, Wang Z. SRT1720 inhibits bladder cancer cell progression by impairing autophagic flux. Biochem Pharmacol 2024; 222:116111. [PMID: 38458329 DOI: 10.1016/j.bcp.2024.116111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/19/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Bladder cancer (BC) is the most common cancer of the urinary tract, with poor survival, high recurrence rates, and lacking of targeted drugs. In this study, we constructed a library to screen compounds inhibiting bladder cancer cells growth. Among them, SRT1720 was identified to inhibit bladder cancer cell proliferation in vitro and in vivo. SRT1720 treatment also suppressed bladder cancer cells migration, invasion and induced apoptosis. Mechanism studies shown that SRT1720 promoted autophagosomes accumulation by inducing early-stage autophagy but disturbed the late-stage of autophagy by blocking fusion of autophagosomes and lysosomes. SRT1720 appears to induce autophagy related proteins expression and alter autophagy-related proteins acetylation to impede the autophagy flux. LAMP2, an important lysosomal associated membrane protein, may mediate SRT1720-inhibited autophagy flux as SRT1720 treatment significantly deacetylated LAMP2 which may influence its activity. Taken together, our results demonstrated that SRT1720 mediated apoptosis and autophagy flux inhibition may be a novel therapeutic strategy for bladder cancer treatment.
Collapse
Affiliation(s)
- Lanlan Li
- Institute of Urology, Key Laboratory of Urological Disease in Gansu Province, Clinical Research Center for Urology in Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China
| | - Shengjun Fu
- Institute of Urology, Key Laboratory of Urological Disease in Gansu Province, Clinical Research Center for Urology in Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China
| | - Jianliang Wang
- Department of Pharmacy, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou 730035, Gansu, China
| | - Jianzhong Lu
- Institute of Urology, Key Laboratory of Urological Disease in Gansu Province, Clinical Research Center for Urology in Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China
| | - Yan Tao
- Institute of Urology, Key Laboratory of Urological Disease in Gansu Province, Clinical Research Center for Urology in Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China
| | - Liangtao Zhao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China
| | - Beitang Fu
- The Fifth Affiliated Hospital of Xinjiang Medical University, Ürümqi 830000, China
| | - Lanpeng Lu
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Caifei Xiang
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Xince Sun
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Shanhui Liu
- Institute of Urology, Key Laboratory of Urological Disease in Gansu Province, Clinical Research Center for Urology in Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China.
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Zhiping Wang
- Institute of Urology, Key Laboratory of Urological Disease in Gansu Province, Clinical Research Center for Urology in Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, Gansu, China.
| |
Collapse
|
12
|
Ferrucci M, Lenzi P, Lazzeri G, Busceti CL, Frati A, Puglisi-Allegra S, Fornai F. Combined light and electron microscopy (CLEM) to quantify methamphetamine-induced alpha-synuclein-related pathology. J Neural Transm (Vienna) 2024; 131:335-358. [PMID: 38367081 PMCID: PMC11016004 DOI: 10.1007/s00702-024-02741-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/09/2024] [Indexed: 02/19/2024]
Abstract
Methamphetamine (METH) produces a cytopathology, which is rather specific within catecholamine neurons both in vitro and ex vivo, in animal models and chronic METH abusers. This led some authors to postulate a sort of parallelism between METH cytopathology and cell damage in Parkinson's disease (PD). In fact, METH increases and aggregates alpha-syn proto-fibrils along with producing spreading of alpha-syn. Although alpha-syn is considered to be the major component of aggregates and inclusions developing within diseased catecholamine neurons including classic Lewy body (LB), at present, no study provided a quantitative assessment of this protein in situ, neither following METH nor in LB occurring in PD. Similarly, no study addressed the quantitative comparison between occurrence of alpha-syn and other key proteins and no investigation measured the protein compared with non-protein structure within catecholamine cytopathology. Therefore, the present study addresses these issues using an oversimplified model consisting of a catecholamine cell line where the novel approach of combined light and electron microscopy (CLEM) was used measuring the amount of alpha-syn, which is lower compared with p62 or poly-ubiquitin within pathological cell domains. The scenario provided by electron microscopy reveals unexpected findings, which are similar to those recently described in the pathology of PD featuring packing of autophagosome-like vesicles and key proteins shuttling autophagy substrates. Remarkably, small seed-like areas, densely packed with p62 molecules attached to poly-ubiquitin within wide vesicular domains occurred. The present data shed new light about quantitative morphometry of catecholamine cell damage in PD and within the addicted brain.
Collapse
Affiliation(s)
- Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Carla L Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy
| | - Alessandro Frati
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy
- Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135, Rome, Italy
| | - Stefano Puglisi-Allegra
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077, Pozzilli, Italy.
| |
Collapse
|
13
|
Waite KA, Vontz G, Lee SY, Roelofs J. Proteasome condensate formation is driven by multivalent interactions with shuttle factors and ubiquitin chains. Proc Natl Acad Sci U S A 2024; 121:e2310756121. [PMID: 38408252 PMCID: PMC10927584 DOI: 10.1073/pnas.2310756121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/26/2024] [Indexed: 02/28/2024] Open
Abstract
Stress conditions can cause the relocalization of proteasomes to condensates in yeast and mammalian cells. The interactions that facilitate the formation of proteasome condensates, however, are unclear. Here, we show that the formation of proteasome condensates in yeast depends on ubiquitin chains together with the proteasome shuttle factors Rad23 and Dsk2. These shuttle factors colocalize to these condensates. Strains deleted for the third shuttle factor gene, DDI1, show proteasome condensates in the absence of cellular stress, consistent with the accumulation of substrates with long K48-linked ubiquitin chains that accumulate in this mutant. We propose a model where the long K48-linked ubiquitin chains function as a scaffold for the ubiquitin-binding domains of the shuttle factors and the proteasome, allowing for the multivalent interactions that further drive condensate formation. Indeed, we determined different intrinsic ubiquitin receptors of the proteasome-Rpn1, Rpn10, and Rpn13-and the Ubl domains of Rad23 and Dsk2 are critical under different condensate-inducing conditions. In all, our data support a model where the cellular accumulation of substrates with long ubiquitin chains, potentially due to reduced cellular energy, allows for proteasome condensate formation. This suggests that proteasome condensates are not simply for proteasome storage, but function to sequester soluble ubiquitinated substrates together with inactive proteasomes.
Collapse
Affiliation(s)
- Kenrick A. Waite
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS66160
| | - Gabrielle Vontz
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS66160
| | - Stella Y. Lee
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS66160
| | - Jeroen Roelofs
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
14
|
Zhou J, Li C, Lu M, Jiang G, Chen S, Li H, Lu K. Pharmacological induction of autophagy reduces inflammation in macrophages by degrading immunoproteasome subunits. PLoS Biol 2024; 22:e3002537. [PMID: 38447109 PMCID: PMC10917451 DOI: 10.1371/journal.pbio.3002537] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Defective autophagy is linked to proinflammatory diseases. However, the mechanisms by which autophagy limits inflammation remain elusive. Here, we found that the pan-FGFR inhibitor LY2874455 efficiently activated autophagy and suppressed expression of proinflammatory factors in macrophages stimulated by lipopolysaccharide (LPS). Multiplex proteomic profiling identified the immunoproteasome, which is a specific isoform of the 20s constitutive proteasome, as a substrate that is degraded by selective autophagy. SQSTM1/p62 was found to be a selective autophagy-related receptor that mediated this degradation. Autophagy deficiency or p62 knockdown blocked the effects of LY2874455, leading to the accumulation of immunoproteasomes and increases in inflammatory reactions. Expression of proinflammatory factors in autophagy-deficient macrophages could be reversed by immunoproteasome inhibitors, confirming the pivotal role of immunoproteasome turnover in the autophagy-mediated suppression on the expression of proinflammatory factors. In mice, LY2874455 protected against LPS-induced acute lung injury and dextran sulfate sodium (DSS)-induced colitis and caused low levels of proinflammatory cytokines and immunoproteasomes. These findings suggested that selective autophagy of the immunoproteasome was a key regulator of signaling via the innate immune system.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Chunxia Li
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Meng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Gaoyue Jiang
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| | - Shanze Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Huihui Li
- West China Second University Hospital, Sichuan University, Chengdu, China
| | - Kefeng Lu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and the Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
15
|
Feng J, Li Y, Zhang Y, Sun S, Sun J, Xu Q, Ji X, Liu Y, Wan Q. Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction. J Transl Med 2024; 22:161. [PMID: 38365674 PMCID: PMC10870664 DOI: 10.1186/s12967-024-04946-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND The autophagy adapter SQSTM1/p62 is crucial for maintaining homeostasis in various organs and cells due to its protein-protein interaction domains and involvement in diverse physiological and pathological processes. Vascular endothelium cells play a unique role in vascular biology and contribute to vascular health. METHODS Using the Cre-loxP system, we generated mice with endothelium cell-specific knockout of p62 mediated by Tek (Tek receptor tyrosine kinase)-cre to investigate the essential role of p62 in the endothelium. In vitro, we employed protein mass spectrometry and IPA to identify differentially expressed proteins upon knockdown of p62. Immunoprecipitation assays were conducted to demonstrate the interaction between p62 and FN1 or LAMC2 in human umbilical vein endothelium cells (HUVECs). Additionally, we identified the degradation pathway of FN1 and LAMC2 using the autophagy inhibitor 3-methyladenine (3-MA) or proteasome inhibitor MG132. Finally, the results of immunoprecipitation demonstrated that the interaction between p62 and LAMC2 was abolished in the PB1 truncation group of p62, while the interaction between p62 and FN1 was abolished in the UBA truncation group of p62. RESULTS Our findings revealed that p62 Endo mice exhibited heart, lung, and kidney fibrosis compared to littermate controls, accompanied by severe cardiac dysfunction. Immunoprecipitation assays provided evidence of p62 acting as an autophagy adapter in the autophagy-lysosome pathway for FN1 and LAMC2 degradation respectively through PB1 and UBA domain with these proteins rather than proteasome system. CONCLUSIONS Our study demonstrates that defects in p62 within endothelium cells induce multi-organ fibrosis and cardiac dysfunction in mice. Our findings indicate that FN1 and LAMC2, as markers of (EndoMT), have detrimental effects on HUVECs and elucidate the autophagy-lysosome degradation mechanism of FN1 and LAMC2.
Collapse
Affiliation(s)
- Jing Feng
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Jinan Central Hospital, Shandong University, Jinan, 250021, Shandong, China
- Qingdao Central Hospital, Shandong University, Qingdao, 266042, Shandong, China
| | - Yan Li
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Yu Zhang
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Shengnan Sun
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Jinan Central Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Jian Sun
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China
| | - Quanlin Xu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China
| | - Xingzhao Ji
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China.
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China.
| | - Qiang Wan
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Jinan Central Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
16
|
Naeem A, Prakash R, Kumari N, Ali Khan M, Quaiyoom Khan A, Uddin S, Verma S, Ab Robertson A, Boltze J, Shadab Raza S. MCC950 reduces autophagy and improves cognitive function by inhibiting NLRP3-dependent neuroinflammation in a rat model of Alzheimer's disease. Brain Behav Immun 2024; 116:70-84. [PMID: 38040385 DOI: 10.1016/j.bbi.2023.11.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/11/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023] Open
Abstract
Alzheimer's disease (AD) is the seventh most common cause of mortality and one of the major causes of disability and vulnerability in the elderly. AD is characterized by gradual cognitive deterioration, the buildup of misfolded amyloid beta (Aβ) peptide, and the generation of neurofibrillary tangles. Despite enormous scientific progress, there is no effective cure for AD. Thus, exploring new treatment options to stop AD or at least slow down its progress is important. In this study, we investigated the potential therapeutic effects of MCC950 on NLRP3-mediated inflammasome-driven inflammation and autophagy in AD. Rats treated with streptozotocin (STZ) exhibited simultaneous activation of the NLRP3 inflammasome and autophagy, as confirmed by Western blot, immunofluorescence, and co-immunoprecipitation analyses. MCC950, a specific NLRP3 inhibitor, was intraperitoneally administered (50 mg/kg body weight) to rats with AD-like symptoms induced by intracerebroventricular STZ injections (3 mg/kg body weight). MCC950 effectively suppressed STZ-induced cognitive impairment and anxiety by inhibiting NLRP3-dependent neuroinflammation. Moreover, our findings indicate that MCC950 exerts neuroprotective effects by attenuating autophagy in neuronal cells. The inhibiting effects of MCC950 on inflammasome activation and autophagy were reproduced in vitro, provding further mechansistic insights into MCC950 therapeutic action. Our findings suggest that MCC950 impedes the progression of AD and may also improve cognitive function through the mitigation of autophagy and NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Abdul Naeem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | - Ravi Prakash
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | - Neha Kumari
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India
| | | | - Abdul Quaiyoom Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Sandeep Verma
- Department of Chemistry, Indian Institute of Technology, Kanpur, UP 208016, India
| | - Avril Ab Robertson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow 226003, India.
| |
Collapse
|
17
|
Zhang C, Jin Y. Ginsenoside Rg5 induces NSCLC cell apoptosis and autophagy through PI3K/Akt/mTOR signaling pathway. Hum Exp Toxicol 2024; 43:9603271241229140. [PMID: 38289222 DOI: 10.1177/09603271241229140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
OBJECTIVE Ginsenoside Rg5 (Rg5) is a minor ginsenoside of ginseng and has a strong anti-tumor potential. This study focused on deciphering the function of Rg5 in non-small cell lung cancer (NSCLC) and investigating its related mechanism. METHODS After treating human NSCLC cell lines (H1650 and A549) and bronchial epithelial cells (BEAS-2B) with increasing concentration of Rg5, cell viability was examined using methyl thiazolyl tetrazolium (MTT) assay. NSCLC cell proliferation and apoptosis were evaluated by colony formation assay and flow cytometry, respectively. The levels of proteins associated with cell cycle progression, cell apoptosis, and autophagy as well as the key markers in the PI3K/Akt/mTOR pathway were measured using western blot. A xenograft nude mouse model was established to explore the function of Rg5 in vivo. RESULTS NSCLC cell viability was dose- and time-dependently suppressed after Rg5 treatment. Rg5 restrained NSCLC cell proliferation by inducing G2/M phase arrest via regulation of cell cycle-related genes including p21, cyclin B1, and Cdc2. Additionally, Rg5 promoted caspase-dependent apoptosis in NSCLC cells by regulating the intrinsic mitochondrial signaling pathway. Rg5 induced autophagy via the regulation of autophagy-related proteins. The in vivo experiments revealed the inhibitory impact of Rg5 on xenograft growth. Rg5 also inactivated the PI3K/Akt/mTOR signaling pathway in NSCLC cells and mouse tumors. CONCLUSION Rg5 induced autophagy and caspase-dependent apoptosis in NSCLC cells by inhibiting the PI3K/Akt/mTOR signaling pathway, suggesting that Rg5 might become a promising and novel anti-tumor agent for the clinical treatment of NSCLC patients.
Collapse
Affiliation(s)
- Caidie Zhang
- Department of Emergency, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yan Jin
- Department of Emergency, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
18
|
Wang T, Zhang Y, Liu Y, Huang Y, Wang W. Amino Acid-Starved Cancer Cells Utilize Macropinocytosis and Ubiquitin-Proteasome System for Nutrient Acquisition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304791. [PMID: 37983609 PMCID: PMC10767443 DOI: 10.1002/advs.202304791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/06/2023] [Indexed: 11/22/2023]
Abstract
To grow in nutrient-deprived tumor microenvironment, cancer cells often internalize and degrade extracellular proteins to refuel intracellular amino acids. However, the nutrient acquisition routes reported by previous studies are mainly restricted in autophagy-lysosomal pathway. It remains largely unknown if other protein degradation systems also contribute to the utilization of extracellular nutrients. Herein, it is demonstrated that under amino acid starvation, extracellular protein internalization through macropinocytosis and protein degradation through ubiquitin-proteasome system are activated as a nutrient supply route, sensitizing cancer cells to proteasome inhibition. By inhibiting both macropinocytosis and ubiquitin-proteasome system, an innovative approach to intensify amino acid starvation for cancer therapy is presented. To maximize therapeutic efficacy and minimize systemic side effects, a pH-responsive polymersome nanocarrier is developed to deliver therapeutic agents specifically to tumor tissues. This nanoparticle system provides an approach to exacerbate amino acid starvation for cancer therapy, which represents a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Tianyi Wang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yaming Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yuwei Liu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yi Huang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| |
Collapse
|
19
|
Hua Z. Deciphering the protein ubiquitylation system in plants. JOURNAL OF EXPERIMENTAL BOTANY 2023; 74:6487-6504. [PMID: 37688404 DOI: 10.1093/jxb/erad354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/07/2023] [Indexed: 09/10/2023]
Abstract
Protein ubiquitylation is a post-translational modification (PTM) process that covalently modifies a protein substrate with either mono-ubiquitin moieties or poly-ubiquitin chains often at the lysine residues. In Arabidopsis, bioinformatic predictions have suggested that over 5% of its proteome constitutes the protein ubiquitylation system. Despite advancements in functional genomic studies in plants, only a small fraction of this bioinformatically predicted system has been functionally characterized. To expand our understanding about the regulatory function of protein ubiquitylation to that rivalling several other major systems, such as transcription regulation and epigenetics, I describe the status, issues, and new approaches of protein ubiquitylation studies in plant biology. I summarize the methods utilized in defining the ubiquitylation machinery by bioinformatics, identifying ubiquitylation substrates by proteomics, and characterizing the ubiquitin E3 ligase-substrate pathways by functional genomics. Based on the functional and evolutionary analyses of the F-box gene superfamily, I propose a deleterious duplication model for the large expansion of this family in plant genomes. Given this model, I present new perspectives of future functional genomic studies on the plant ubiquitylation system to focus on core and active groups of ubiquitin E3 ligase genes.
Collapse
Affiliation(s)
- Zhihua Hua
- Department of Environmental and Plant Biology, Ohio University, Athens, OH 45701, USA
- Interdisciplinary Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
20
|
Langer HT, Taylor SR, Ahmed M, Perrier T, Ahmed T, Goncalves MD. The proteasome regulates body weight and systemic nutrient metabolism during fasting. Am J Physiol Endocrinol Metab 2023; 325:E500-E512. [PMID: 37672249 PMCID: PMC10864006 DOI: 10.1152/ajpendo.00069.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023]
Abstract
The ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway are the primary means of degradation in mammalian tissues. We sought to determine the individual contribution of the UPS and autophagy to tissue catabolism during fasting. Mice were overnight fasted for 15 h before regaining food access ("Fed" group, n = 6) or continuing to fast ("Fast" group, n = 7) for 3 h. In addition, to investigate the effects of autophagy on systemic metabolism and tissue degradation, one group of mice was fasted for 18 h and treated with chloroquine ("Fast + CLQ" group, n = 7) and a fourth group of mice was treated with bortezomib ("Fast + Bort" group, n = 7) to assess the contribution of the UPS. Body weight, tissue weight, circulating hormones and metabolites, intracellular signaling pathways, and protein synthesis were investigated. Fasting induced the loss of body weight, liver mass, and white adipose tissue in the Fast and the Fast + CLQ group, whereas the Fast + Bort group maintained tissue and body weight. Fasting reduced glucose and increased β hydroxybutyrate in the circulation of all mice. Both changes were most profound in the Fast + Bort group compared with the other fasting conditions. Molecular signaling indicated a successful inhibition of hepatic UPS with bortezomib and an upregulation of the PI3K/AKT/mTOR pathway. The latter was further supported by an increase in hepatic protein synthesis with bortezomib. Inhibition of the UPS through bortezomib blocks body weight loss and tissue catabolism during an acute overnight fast in mice. The effects were likely mediated through a combined effect of the drug on biomolecule degradation and synthesis.NEW & NOTEWORTHY Bortezomib treatment prevents tissue and body weight loss during fasting. The loss of proteasome activity with bortezomib exacerbates fasting-induced ketogenesis. During fasting, bortezomib increases AMPK and PI3K/AKT signaling in the liver, which promotes protein synthesis.
Collapse
Affiliation(s)
- Henning Tim Langer
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States
| | - Samuel R Taylor
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States
| | - Mujmmail Ahmed
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States
| | - Tiffany Perrier
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States
| | - Tanvir Ahmed
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States
| | - Marcus D Goncalves
- Department of Medicine, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
21
|
Livneh I, Cohen-Kaplan V, Fabre B, Abramovitch I, Lulu C, Nataraj NB, Lazar I, Ziv T, Yarden Y, Zohar Y, Gottlieb E, Ciechanover A. Regulation of nucleo-cytosolic 26S proteasome translocation by aromatic amino acids via mTOR is essential for cell survival under stress. Mol Cell 2023; 83:3333-3346.e5. [PMID: 37738964 DOI: 10.1016/j.molcel.2023.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 09/24/2023]
Abstract
The proteasome is responsible for removal of ubiquitinated proteins. Although several aspects of its regulation (e.g., assembly, composition, and post-translational modifications) have been unraveled, studying its adaptive compartmentalization in response to stress is just starting to emerge. We found that following amino acid starvation, the proteasome is translocated from its large nuclear pool to the cytoplasm-a response regulated by newly identified mTOR-agonistic amino acids-Tyr, Trp, and Phe (YWF). YWF relay their signal upstream of mTOR through Sestrin3 by disrupting its interaction with the GATOR2 complex. The triad activates mTOR toward its downstream substrates p62 and transcription factor EB (TFEB), affecting both proteasomal and autophagic activities. Proteasome translocation stimulates cytosolic proteolysis which replenishes amino acids, thus enabling cell survival. In contrast, nuclear sequestration of the proteasome following mTOR activation by YWF inhibits this proteolytic adaptive mechanism, leading to cell death, which establishes this newly identified pathway as a key stress-coping mechanism.
Collapse
Affiliation(s)
- Ido Livneh
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel; Institute of Pathology, Rambam Health Care Campus, Haifa 3109601, Israel.
| | - Victoria Cohen-Kaplan
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Bertrand Fabre
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Ifat Abramovitch
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Chen Lulu
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | | | - Ikrame Lazar
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Tamar Ziv
- Smoler Proteomic Center, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yaniv Zohar
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel; Institute of Pathology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Eyal Gottlieb
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Aaron Ciechanover
- The Rappaport Faculty of Medicine and Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa 3109601, Israel.
| |
Collapse
|
22
|
Zhang C, Xu T, Ji K, Cao S, Cao Y, Ai J, Jing L, Sun JH. An integrative analysis reveals the prognostic value and potential functions of PSMD11 in hepatocellular carcinoma. Mol Carcinog 2023; 62:1355-1368. [PMID: 37212487 DOI: 10.1002/mc.23568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/23/2023]
Abstract
The global burden of hepatocellular carcinoma (HCC) as a preeminent etiology of cancer-related mortalities sheds light on the imperative necessity for a more profound comprehension of its fundamental biological mechanisms. In this context, the precise function of the 26S proteasome non-ATPase regulatory subunit 11 (PSMD11) in HCC remains equivocal. To address this vital knowledge gap, we interrogated the cancer genome atlas, genotype-tissue expression, International cancer genome consortium, gene expression omnibus, the cancer cell line encyclopedia, and tumor immune single-cell hub databases to evaluate the expression pattern of PSMD11, further confirmed by reverse-transcription quantitative polymerase chain reaction (RT-qPCR) in LO2, MHCC-97H, HepG2, and SMMC7721 cell lines. Additionally, we meticulously assessed the clinical significance and prognostic value of PSMD11, while also exploring its potential molecular mechanisms in HCC. Our findings demonstrated that PSMD11 was highly expressed in HCC tissues, correlating with pathologic stage and histologic grade, thereby conferring a poor prognosis. Mechanistically, PSMD11 appears to exert its tumorigenic effects through the modulation of tumor metabolism-related pathways. Impressively, low PSMD11 expression was associated with increased immune effector cell infiltration, heightened responsiveness to molecular targeted drugs such as dasatinib, erlotinib, gefitinib, and imatinib, as well as reduced somatic mutation rate. Additionally, we demonstrated that PSMD11 might modulate HCC development through intricate interactions with cuproptosis-related genes ATP7A, DLAT, and PDHA1. Our comprehensive analyses collectively suggest that PSMD11 represents a promising therapeutic target in HCC.
Collapse
Affiliation(s)
- Cong Zhang
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Tiantian Xu
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Kun Ji
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Shoujin Cao
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Yunbo Cao
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Jing Ai
- Department of Ophthalmology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Li Jing
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
| | - Jun-Hui Sun
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang Province, China
- Zhejiang University Cancer Center, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Ma J, Zhou W, Yuan Y, Wang B, Meng X. PSMD12 interacts with CDKN3 and facilitates pancreatic cancer progression. Cancer Gene Ther 2023; 30:1072-1083. [PMID: 37037907 DOI: 10.1038/s41417-023-00609-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 04/12/2023]
Abstract
Proteasome 26S subunit, non-ATPase 12 (PSMD12) genes have been implicated in several types of malignancies but the role of PSMD12 in pancreatic cancer (PC) remains elusive. Bioinformatics analysis showed that PSMD12 was highly expressed in PC patients and was associated with shorter overall survival. PSMD12 was also shown to be highly expressed in PC tissues and cell lines. Upregulated PSMD12 showed enhanced cell viability, increased colony formation rate and upregulated levels of PCNA and c-Myc, while the inhibition of PSMD12 abated these levels. PSMD12 knockdown promoted cell apoptosis. The results of xenografts in nude mice confirmed that PSMD12 promoted PC tumor growth in vivo. Protein‒protein interaction network and functional enrichment analyses implied that PSMD12 may have a connection with cyclin-dependent kinase inhibitor 3 (CDKN3). Co‑immunoprecipitation and western blot results confirmed that PSMD12 could interact with and abate the ubiquitination level of CDKN3, thus stabilizing the CDKN3 protein. Rescue assays showed that PSMD12 overexpression caused cell proliferation and that knockdown-induced cell apoptosis could be reversed by CDKN3 regulation. This work reveals the essential roles of PSMD12 in the proliferation and apoptosis of PC development. PSMD12 may regulate CDKN3 expression by interacting with and abating the ubiquitination level of CDKN3, thereby participating in the malignant behavior of PC.
Collapse
Affiliation(s)
- Jia Ma
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wenyang Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yifeng Yuan
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Baosheng Wang
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiangpeng Meng
- Pancreatic Endocrinology Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
24
|
Waite KA, Vontz G, Lee SY, Roelofs J. Proteasome condensate formation is driven by multivalent interactions with shuttle factors and K48-linked ubiquitin chains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.25.546446. [PMID: 37425862 PMCID: PMC10326979 DOI: 10.1101/2023.06.25.546446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Stress conditions can cause the relocalization of proteasomes to condensates in yeast and mammalian cells. The interactions that facilitate the formation of proteasome condensates, however, are unclear. Here, we show that the formation of proteasome condensates in yeast depends on long K48-linked ubiquitin chains together with the proteasome shuttle factors Rad23 and Dsk2. These shuttle factors colocalize to these condensates. Strains deleted for the third shuttle factor gene, DDI1 , show proteasome condensates in the absence of cellular stress, consistent with the accumulation of substrates with long K48-linked ubiquitin chains that accumulate in this mutant. We propose a model where the long K48-linked ubiquitin chains function as a scaffold for the ubiquitin binding domains of the shuttle factors and the proteasome, allowing for the multivalent interactions that further drive condensate formation. Indeed, we determined different intrinsic ubiquitin receptors of the proteasome (Rpn1, Rpn10, and Rpn13) are critical under different condensate inducing conditions. In all, our data support a model where the cellular accumulation of substrates with long ubiquitin chains, potentially due to reduced cellular energy, allows for proteasome condensate formation. This suggests that proteasome condensates are not simply for proteasome storage, but function to sequester soluble ubiquitinated substrates together with inactive proteasomes. Significance Stress conditions can cause the relocalization of proteasomes to condensates in yeast as well as mammalian cells. Our work shows that the formation of proteasome condensates in yeast depends on long K48-linked ubiquitin chains, the proteasome binding shuttle factors Rad23 and Dsk2 and proteasome intrinsic ubiquitin receptors. Here, different receptors are critical for different condensate inducers. These results indicate distinct condensates can form with specific functionality. Our identification of key factors involved in the process is crucial for understanding the function of proteasome relocalization to condensates. We propose that cellular accumulation of substrates with long ubiquitin chains results in the formation of condensates comprising those ubiquitinated substrates, proteasomes, and proteasome shuttle factors, where the ubiquitin chains serve as the scaffold for condensate formation.
Collapse
|
25
|
Hanson S, Dharan A, P. V. J, Pal S, Nair BG, Kar R, Mishra N. Paraptosis: a unique cell death mode for targeting cancer. Front Pharmacol 2023; 14:1159409. [PMID: 37397502 PMCID: PMC10308048 DOI: 10.3389/fphar.2023.1159409] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/15/2023] [Indexed: 07/04/2023] Open
Abstract
Programmed cell death (PCD) is the universal process that maintains cellular homeostasis and regulates all living systems' development, health and disease. Out of all, apoptosis is one of the major PCDs that was found to play a crucial role in many disease conditions, including cancer. The cancer cells acquire the ability to escape apoptotic cell death, thereby increasing their resistance towards current therapies. This issue has led to the need to search for alternate forms of programmed cell death mechanisms. Paraptosis is an alternative cell death pathway characterized by vacuolation and damage to the endoplasmic reticulum and mitochondria. Many natural compounds and metallic complexes have been reported to induce paraptosis in cancer cell lines. Since the morphological and biochemical features of paraptosis are much different from apoptosis and other alternate PCDs, it is crucial to understand the different modulators governing it. In this review, we have highlighted the factors that trigger paraptosis and the role of specific modulators in mediating this alternative cell death pathway. Recent findings include the role of paraptosis in inducing anti-tumour T-cell immunity and other immunogenic responses against cancer. A significant role played by paraptosis in cancer has also scaled its importance in knowing its mechanism. The study of paraptosis in xenograft mice, zebrafish model, 3D cultures, and novel paraptosis-based prognostic model for low-grade glioma patients have led to the broad aspect and its potential involvement in the field of cancer therapy. The co-occurrence of different modes of cell death with photodynamic therapy and other combinatorial treatments in the tumour microenvironment are also summarized here. Finally, the growth, challenges, and future perspectives of paraptosis research in cancer are discussed in this review. Understanding this unique PCD pathway would help to develop potential therapy and combat chemo-resistance in various cancer.
Collapse
Affiliation(s)
- Sweata Hanson
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Aiswarya Dharan
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Jinsha P. V.
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Sanjay Pal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Bipin G. Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Rekha Kar
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, United States
| | - Nandita Mishra
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| |
Collapse
|
26
|
Raffeiner M, Zhu S, González-Fuente M, Üstün S. Interplay between autophagy and proteasome during protein turnover. TRENDS IN PLANT SCIENCE 2023; 28:698-714. [PMID: 36801193 DOI: 10.1016/j.tplants.2023.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 05/13/2023]
Abstract
Protein homeostasis is epitomized by an equilibrium between protein biosynthesis and degradation: the 'life and death' of proteins. Approximately one-third of newly synthesized proteins are degraded. As such, protein turnover is required to maintain cellular integrity and survival. Autophagy and the ubiquitin-proteasome system (UPS) are the two principal degradation pathways in eukaryotes. Both pathways orchestrate many cellular processes during development and upon environmental stimuli. Ubiquitination of degradation targets is used as a 'death' signal by both processes. Recent findings revealed a direct functional link between both pathways. Here, we summarize key findings in the field of protein homeostasis, with an emphasis on the newly revealed crosstalk between both degradation machineries and how it is decided which pathway facilitates target degradation.
Collapse
Affiliation(s)
- Margot Raffeiner
- Eberhard-Karls-Universität Tübingen, Zentrum für Molekular Biologie der Pflanzen, 72076 Tübingen, Germany; Faculty of Biology & Biotechnology, Ruhr-University of Bochum, 44780 Bochum, Germany
| | - Shanshuo Zhu
- Eberhard-Karls-Universität Tübingen, Zentrum für Molekular Biologie der Pflanzen, 72076 Tübingen, Germany; Faculty of Biology & Biotechnology, Ruhr-University of Bochum, 44780 Bochum, Germany
| | - Manuel González-Fuente
- Eberhard-Karls-Universität Tübingen, Zentrum für Molekular Biologie der Pflanzen, 72076 Tübingen, Germany; Faculty of Biology & Biotechnology, Ruhr-University of Bochum, 44780 Bochum, Germany
| | - Suayib Üstün
- Eberhard-Karls-Universität Tübingen, Zentrum für Molekular Biologie der Pflanzen, 72076 Tübingen, Germany; Faculty of Biology & Biotechnology, Ruhr-University of Bochum, 44780 Bochum, Germany.
| |
Collapse
|
27
|
El-Deeb AM, Mohamed AF, El-Yamany MF, El-Tanbouly DM. Novel trajectories of the NK1R antagonist aprepitant in rotenone-induced Parkinsonism-like symptoms in rats: Involvement of ERK5/KLF4/p62/Nrf2 signaling axis. Chem Biol Interact 2023; 380:110562. [PMID: 37224993 DOI: 10.1016/j.cbi.2023.110562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/09/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
Regulation of the interplay between autophagy and oxidative stress is vital in maintaining neuronal homeostasis during neurotoxicity. The interesting involvement of NK1 receptor (NK1R) in neurodegeneration has highlighted the value of investigating the neuroprotective effect of aprepitant (Aprep), an NK1R antagonist in Parkinson's disease (PD). This study was conducted to disclose Aprep's ability to modulate extracellular signal-regulated kinase 5/Krüppel-like factor 4 (ERK5/KLF4) cue as molecular signaling implicated in regulating autophagy and redox signaling in response to rotenone neurotoxicity. Rotenone (1.5 mg/kg) was administered on alternate days, and rats were given Aprep simultaneously with or without PD98059, an ERK inhibitor, for 21 days. Aprep ameliorated motor deficits as verified by restored histological features, and intact neurons count in SN and striata along with tyrosine hydroxylase immunoreactivity in SN. The molecular signaling of Aprep was illustrated by the expression of KLF4 following the phosphorylation of its upstream target, ERK5. Nuclear factor erythroid 2-related factor 2 (Nrf2) was up-regulated, shifting the oxidant/antioxidant balance towards the antioxidant side, as evidenced by elevated GSH and suppressed MDA levels. In parallel, Aprep noticeably reduced phosphorylated α-synuclein aggregates due to autophagy induction as emphasized by marked LC3II/LC3I elevation and p62 level reduction. These effects were diminished upon PD98059 pre-administration. In conclusion, Aprep showed neuroprotective effects against rotenone-induced PD, which may be partially attributed to the activation of the ERK5/KLF4 signaling pathway. It modulated p62-mediated autophagy and Nrf2 axis which act cooperatively to counter rotenone-associated neurotoxicity pointing to Aprep's prospect as a curious candidate in PD research.
Collapse
Affiliation(s)
- Asmaa M El-Deeb
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt.
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| |
Collapse
|
28
|
Hao X, Sun J, Zhong L, Baudry M, Bi X. UBE3A deficiency-induced autophagy is associated with activation of AMPK-ULK1 and p53 pathways. Exp Neurol 2023; 363:114358. [PMID: 36849003 PMCID: PMC10073344 DOI: 10.1016/j.expneurol.2023.114358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/03/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Angelman Syndrome (AS) is a neurodevelopmental disorder caused by deficiency of the maternally expressed UBE3A gene. The UBE3A proteins functions both as an E3 ligase in the ubiquitin-proteasome system (UPS), and as a transcriptional co-activator for steroid hormone receptors. Here we investigated the effects of UBE3A deficiency on autophagy in the cerebellum of AS mice and in COS1 cells. Numbers and size of LC3- and LAMP2-immunopositive puncta were increased in cerebellar Purkinje cells of AS mice, as compared to wildtype mice. Western blot analysis showed an increase in the conversion of LC3I to LC3II in AS mice, as expected from increased autophagy. Levels of active AMPK and of one of its substrates, ULK1, a factor involved in autophagy initiation, were also increased. Colocalization of LC3 with LAMP2 was increased and p62 levels were decreased, indicating an increase in autophagy flux. UBE3A deficiency was also associated with reduced levels of phosphorylated p53 in the cytosol and increased levels in nuclei, which favors autophagy induction. UBE3A siRNA knockdown in COS-1 cells resulted in increased size and intensity of LC3-immunopositive puncta and increased the LC3 II/I ratio, as compared to control siRNA-treated cells, confirming the results found in the cerebellum of AS mice. These results indicate that UBE3A deficiency enhances autophagic activity through activation of the AMPK-ULK1 pathway and alterations in p53.
Collapse
Affiliation(s)
- Xiaoning Hao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Li Zhong
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA.
| |
Collapse
|
29
|
Shames SR. Eat or Be Eaten: Strategies Used by Legionella to Acquire Host-Derived Nutrients and Evade Lysosomal Degradation. Infect Immun 2023; 91:e0044122. [PMID: 36912646 PMCID: PMC10112212 DOI: 10.1128/iai.00441-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023] Open
Abstract
To replicate within host cells, bacterial pathogens must acquire host-derived nutrients while avoiding degradative antimicrobial pathways. Fundamental insights into bacterial pathogenicity have been revealed by bacteria of the genus Legionella, which naturally parasitize free-living protozoa by establishing a membrane-bound replicative niche termed the Legionella-containing vacuole (LCV). Biogenesis of the LCV and intracellular replication rely on rapid evasion of the endocytic pathway and acquisition of host-derived nutrients, much of which is mediated by bacterial effector proteins translocated into host cells by a Dot/Icm type IV secretion system. Billions of years of co-evolution with eukaryotic hosts and broad host tropism have resulted in expansion of the Legionella genome to accommodate a massive repertoire of effector proteins that promote LCV biogenesis, safeguard the LCV from endolysosomal maturation, and mediate the acquisition of host nutrients. This minireview is focused on the mechanisms by which an ancient intracellular pathogen leverages effector proteins and hijacks host cell biology to obtain essential host-derived nutrients and prevent lysosomal degradation.
Collapse
Affiliation(s)
- Stephanie R. Shames
- Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
30
|
Wang L, Klionsky DJ, Shen HM. The emerging mechanisms and functions of microautophagy. Nat Rev Mol Cell Biol 2023; 24:186-203. [PMID: 36097284 DOI: 10.1038/s41580-022-00529-z] [Citation(s) in RCA: 172] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 02/08/2023]
Abstract
'Autophagy' refers to an evolutionarily conserved process through which cellular contents, such as damaged organelles and protein aggregates, are delivered to lysosomes for degradation. Different forms of autophagy have been described on the basis of the nature of the cargoes and the means used to deliver them to lysosomes. At present, the prevailing categories of autophagy in mammalian cells are macroautophagy, microautophagy and chaperone-mediated autophagy. The molecular mechanisms and biological functions of macroautophagy and chaperone-mediated autophagy have been extensively studied, but microautophagy has received much less attention. In recent years, there has been a growth in research on microautophagy, first in yeast and then in mammalian cells. Here we review this form of autophagy, focusing on selective forms of microautophagy. We also discuss the upstream regulatory mechanisms, the crosstalk between macroautophagy and microautophagy, and the functional implications of microautophagy in diseases such as cancer and neurodegenerative disorders in humans. Future research into microautophagy will provide opportunities to develop novel interventional strategies for autophagy- and lysosome-related diseases.
Collapse
Affiliation(s)
- Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| | - Han-Ming Shen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
31
|
Passively-targeted mitochondrial tungsten-based nanodots for efficient acute kidney injury treatment. Bioact Mater 2023; 21:381-393. [PMID: 36185743 PMCID: PMC9483742 DOI: 10.1016/j.bioactmat.2022.08.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 12/25/2022] Open
Abstract
Acute kidney injury (AKI) can lead to loss of kidney function and a substantial increase in mortality. The burst of reactive oxygen species (ROS) plays a key role in the pathological progression of AKI. Mitochondrial-targeted antioxidant therapy is very promising because mitochondria are the main source of ROS in AKI. Antioxidant nanodrugs with actively targeted mitochondria have achieved encouraging success in many oxidative stress-induced diseases. However, most strategies to actively target mitochondria make the size of nanodrugs too large to pass through the glomerular system to reach the renal tubules, the main damage site of AKI. Here, an ultra-small Tungsten-based nanodots (TWNDs) with strong ROS scavenging can be very effective for treatment of AKI. TWNDs can reach the tubular site after crossing the glomerular barrier, and enter the mitochondria of the renal tubule without resorting to complex active targeting strategies. To our knowledge, this is the first time that ultra-small negatively charged nanodots can be used to passively target mitochondrial therapy for AKI. Through in-depth study of the therapeutic mechanism, such passive mitochondria-targeted TWNDs are highly effective in protecting mitochondria by reducing mitochondrial ROS and increasing mitophagy. In addition, TWNDs can also reduce the infiltration of inflammatory cells. This work provides a new way to passively target mitochondria for AKI, and give inspiration for the treatment of many major diseases closely related to mitochondria, such as myocardial infarction and cerebral infarction. Mitochondrial-targeted antioxidant therapy is a very promising treatment for AKI. TWNDs have a strong ability to scavenge ROS, and their small size allows them to cross the glomerular filtration barrier. TWNDs passively target the mitochondria of renal tubular cells and are highly effective in restoring renal function.
Collapse
|
32
|
Mauthe M, Kampinga HH, Hipp MS, Reggiori F. Digest it all: the lysosomal turnover of cytoplasmic aggregates. Trends Biochem Sci 2023; 48:216-228. [PMID: 36280494 DOI: 10.1016/j.tibs.2022.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 02/18/2023]
Abstract
Aggrephagy describes the selective lysosomal transport and turnover of cytoplasmic protein aggregates by macro-autophagy. In this process, protein aggregates and conglomerates are polyubiquitinated and then sequestered by autophagosomes. Soluble selective autophagy receptors (SARs) are central to aggrephagy and physically bind to both ubiquitin and the autophagy machinery, thus linking the cargo to the forming autophagosomal membrane. Because the accumulation of protein aggregates is associated with cytotoxicity in several diseases, a better molecular understanding of aggrephagy might provide a conceptual framework to develop therapeutic strategies aimed at delaying the onset of these pathologies by preventing the buildup of potentially toxic aggregates. We review recent advances in our knowledge about the mechanism of aggrephagy.
Collapse
Affiliation(s)
- Mario Mauthe
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harm H Kampinga
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark S Hipp
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark.
| |
Collapse
|
33
|
Zakluta AS, Shilova VY, Zatsepina OG. The Effect of the Knockout of Major Transsulfuration Genes on the Pattern of Protein Synthesis in D. melanogaster. Mol Biol 2023. [DOI: 10.1134/s0026893323010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
34
|
Zhao HY, Xin M, Zhang SQ. Progress of small molecules for targeted protein degradation: PROTACs and other technologies. Drug Dev Res 2023; 84:337-394. [PMID: 36606428 DOI: 10.1002/ddr.22026] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/01/2022] [Accepted: 12/17/2022] [Indexed: 01/07/2023]
Abstract
Recent years have witnessed the rapid development of targeted protein degradation (TPD), especially proteolysis targeting chimeras. These degraders have manifested many advantages over small molecule inhibitors. To date, a huge number of degraders have been excavated against over 70 disease-related targets. In particular, degraders against estrogen receptor and androgen receptor have crowded into phase II clinical trial. TPD technologies largely expand the scope of druggable targets, and provide powerful tools for addressing intractable problems that can not be tackled by traditional small molecule inhibitors. In this review, we mainly focus on the structures and biological activities of small molecule degraders as well as the elucidation of mechanisms of emerging TPD technologies. We also propose the challenges that exist in the TPD field at present.
Collapse
Affiliation(s)
- Hong-Yi Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
35
|
Aepala MR, Peiris MN, Jiang Z, Yang W, Meyer AN, Donoghue DJ. Nefarious NTRK oncogenic fusions in pediatric sarcomas: Too many to Trk. Cytokine Growth Factor Rev 2022; 68:93-106. [PMID: 36153202 DOI: 10.1016/j.cytogfr.2022.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 01/30/2023]
Abstract
Neurotrophic Tyrosine Receptor Kinase (NTRK) genes undergo chromosomal translocations to create novel open reading frames coding for oncogenic fusion proteins; the N-terminal portion, donated by various partner genes, becomes fused to the tyrosine kinase domain of either NTRK1, NTRK2, or NTRK3. NTRK fusion proteins have been identified as driver oncogenes in a wide variety of tumors over the past three decades, including Pediatric Gliomas, Papillary Thyroid Carcinoma, Spitzoid Neoplasms, Glioblastoma, and additional tumors. Importantly, NTRK fusions function as drivers of pediatric sarcomas, accounting for approximately 15% of childhood cancers including Infantile Fibrosarcoma (IFS), a subset of pediatric soft tissue sarcoma (STS). While tyrosine kinase inhibitors (TKIs), such as larotrectinib and entrectinib, have demonstrated profound results against NTRK fusion-positive cancers, acquired resistance to these TKIs has resulted in the formation of gatekeeper, solvent-front, and compound mutations. We present a comprehensive compilation of oncogenic fusions involving NTRKs focusing specifically on pediatric STS, examining their biological signaling pathways and mechanisms of activation. The importance of an obligatory dimerization or multimerization domain, invariably donated by the N-terminal fusion partner, is discussed using characteristic fusions that occur in pediatric sarcomas. In addition, examples are presented of oncogenic fusion proteins in which the N-terminal partners may contribute additional biological activities beyond an oligomerization domain. Lastly, therapeutic approaches to the treatment of pediatric sarcoma will be presented, using first generation and second-generation agents such as selitrectinib and repotrectinib.
Collapse
Affiliation(s)
- Megha R Aepala
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - Malalage N Peiris
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - Zian Jiang
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - Wei Yang
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - April N Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0367, USA; UCSD Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, USA.
| |
Collapse
|
36
|
Gonzalez-Santamarta M, Bouvier C, Rodriguez MS, Xolalpa W. Ubiquitin-chains dynamics and its role regulating crucial cellular processes. Semin Cell Dev Biol 2022; 132:155-170. [PMID: 34895814 DOI: 10.1016/j.semcdb.2021.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
The proteome adapts to multiple situations occurring along the life of the cell. To face these continuous changes, the cell uses posttranslational modifications (PTMs) to control the localization, association with multiple partners, stability, and activity of protein targets. One of the most dynamic protein involved in PTMs is Ubiquitin (Ub). Together with other members of the same family, known as Ubiquitin-like (UbL) proteins, Ub rebuilds the architecture of a protein in a few minutes to change its properties in a very efficient way. This capacity of Ub and UbL is in part due to their potential to form complex architectures when attached to target proteins or when forming Ub chains. The highly dynamic formation and remodeling of Ub chains is regulated by the action of conjugating and deconjugating enzymes that determine, in due time, the correct chain architecture for a particular cellular function. Chain remodeling occurs in response to physiologic stimuli but also in pathologic situations. Here, we illustrate well-documented cases of chain remodeling during DNA repair, activation of the NF-κB pathway and autophagy, as examples of this dynamic regulation. The crucial role of enzymes and cofactors regulating chain remodeling is discussed.
Collapse
Affiliation(s)
- Maria Gonzalez-Santamarta
- Laboratoire de Chimie de Coordination (LCC) - UPR 8241 CNRS, and UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France.
| | - Corentin Bouvier
- Laboratoire de Chimie de Coordination (LCC) - UPR 8241 CNRS, and UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France.
| | - Manuel S Rodriguez
- Laboratoire de Chimie de Coordination (LCC) - UPR 8241 CNRS, and UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31400 Toulouse, France.
| | - Wendy Xolalpa
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, 62250 Cuernavaca, Morelos, Mexico.
| |
Collapse
|
37
|
Kocaturk NM, Peker N, Eberhart K, Akkoc Y, Deveci G, Dengjel J, Gozuacik D. Novel protein complexes containing autophagy and UPS components regulate proteasome-dependent PARK2 recruitment onto mitochondria and PARK2-PARK6 activity during mitophagy. Cell Death Dis 2022; 13:947. [PMID: 36357363 PMCID: PMC9649694 DOI: 10.1038/s41419-022-05339-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 08/31/2022] [Accepted: 10/12/2022] [Indexed: 11/11/2022]
Abstract
Autophagy is an evolutionarily conserved eukaryotic cellular mechanism through which cytosolic fragments, misfolded/aggregated proteins and organelles are degraded and recycled. Priming of mitochondria through ubiquitylation is required for the clearance the organelle by autophagy (mitophagy). Familial Parkinson's Disease-related proteins, including the E3-ligase PARK2 (PARKIN) and the serine/threonine kinase PARK6 (PINK1) control these ubiquitylation reactions and contribute to the regulation of mitophagy. Here we describe, novel protein complexes containing autophagy protein ATG5 and ubiquitin-proteasome system (UPS) components. We discovered that ATG5 interacts with PSMA7 and PARK2 upon mitochondrial stress. Results suggest that all three proteins translocate mitochondria and involve in protein complexes containing autophagy, UPS and mitophagy proteins. Interestingly, PARK2 and ATG5 recruitment onto mitochondria requires proteasome components PSMA7 and PSMB5. Strikingly, we discovered that subunit of 20 S proteasome, PSMA7, is required for the progression of PARK2-PARK6-mediated mitophagy and the proteasome activity following mitochondrial stress. Our results demonstrate direct, dynamic and functional interactions between autophagy and UPS components that contribute to the regulation of mitophagy.
Collapse
Affiliation(s)
- Nur Mehpare Kocaturk
- SUNUM Nanotechnology Research and Application Center, Istanbul, Turkey
- School of Life Sciences, University of Dundee, Dundee, UK
| | - Nesibe Peker
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Karin Eberhart
- SUNUM Nanotechnology Research and Application Center, Istanbul, Turkey
| | - Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Gamze Deveci
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | | | - Devrim Gozuacik
- SUNUM Nanotechnology Research and Application Center, Istanbul, Turkey.
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
- Koç University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
38
|
Sparks A, Kelly CJ, Saville MK. Ubiquitin receptors play redundant roles in the proteasomal degradation of the p53 repressor MDM2. FEBS Lett 2022; 596:2746-2767. [PMID: 35735670 PMCID: PMC9796813 DOI: 10.1002/1873-3468.14436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 01/07/2023]
Abstract
Much remains to be determined about the participation of ubiquitin receptors in proteasomal degradation and their potential as therapeutic targets. Suppression of the ubiquitin receptor S5A/PSMD4/hRpn10 alone stabilises p53/TP53 but not the key p53 repressor MDM2. Here, we observed S5A and the ubiquitin receptors ADRM1/PSMD16/hRpn13 and RAD23A and B functionally overlap in MDM2 degradation. We provide further evidence that degradation of only a subset of ubiquitinated proteins is sensitive to S5A knockdown because ubiquitin receptor redundancy is commonplace. p53 can be upregulated by S5A modulation while degradation of substrates with redundant receptors is maintained. Our observations and analysis of Cancer Dependency Map (DepMap) screens show S5A depletion/loss substantially reduces cancer cell line viability. This and selective S5A dependency of proteasomal substrates make S5A a target of interest for cancer therapy.
Collapse
Affiliation(s)
| | - Christopher J. Kelly
- School of MedicineUniversity of DundeeUK,Institute of Infection, Immunity and InflammationUniversity of GlasgowUK
| | - Mark K. Saville
- School of MedicineUniversity of DundeeUK,Silver River EditingDundeeUK
| |
Collapse
|
39
|
Actin dynamics in protein homeostasis. Biosci Rep 2022; 42:231720. [PMID: 36043949 PMCID: PMC9469105 DOI: 10.1042/bsr20210848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/22/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Cell homeostasis is maintained in all organisms by the constant adjustment of cell constituents and organisation to account for environmental context. Fine-tuning of the optimal balance of proteins for the conditions, or protein homeostasis, is critical to maintaining cell homeostasis. Actin, a major constituent of the cytoskeleton, forms many different structures which are acutely sensitive to the cell environment. Furthermore, actin structures interact with and are critically important for the function and regulation of multiple factors involved with mRNA and protein production and degradation, and protein regulation. Altogether, actin is a key, if often overlooked, regulator of protein homeostasis across eukaryotes. In this review, we highlight these roles and how they are altered following cell stress, from mRNA transcription to protein degradation.
Collapse
|
40
|
Waite KA, Roelofs J. Proteasome granule formation is regulated through mitochondrial respiration and kinase signaling. J Cell Sci 2022; 135:jcs259778. [PMID: 35975718 PMCID: PMC9482347 DOI: 10.1242/jcs.259778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/03/2022] [Indexed: 11/20/2022] Open
Abstract
In the yeast Saccharomyces cerevisiae, proteasomes are enriched in cell nuclei, in which they execute important cellular functions. Nutrient stress can change this localization, indicating that proteasomes respond to the metabolic state of the cell. However, the signals that connect these processes remain poorly understood. Carbon starvation triggers a reversible translocation of proteasomes to cytosolic condensates known as proteasome storage granules. Surprisingly, we observed strongly reduced levels of proteasome granules when cells had active cellular respiration prior to starvation. This suggests that the mitochondrial activity of cells is a determining factor in the response of proteasomes to carbon starvation. Consistent with this, upon inhibition of mitochondrial function, we observed that proteasomes relocalize to granules. These links between proteasomes and metabolism involve specific signaling pathways, as we identified a mitogen-activated protein kinase (MAPK) cascade that is critical to the formation of proteasome granules after respiratory growth but not following glycolytic growth. Furthermore, the yeast homolog of AMP kinase, Snf1, is important for proteasome granule formation induced by mitochondrial inhibitors, but it is dispensable for granule formation following carbon starvation. We propose a model in which mitochondrial activity promotes nuclear localization of the proteasome. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Jeroen Roelofs
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., HLSIC 1077, Kansas City, KS 66160-7421, USA
| |
Collapse
|
41
|
Pu M, Tai Y, Yuan L, Zhang Y, Guo H, Hao Z, Chen J, Qi X, Wang G, Tao Z, Ren J. The contribution of proteasomal impairment to autophagy activation by C9orf72 poly-GA aggregates. Cell Mol Life Sci 2022; 79:501. [PMID: 36036324 DOI: 10.1007/s00018-022-04518-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/18/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Poly-GA, a dipeptide repeat protein unconventionally translated from GGGGCC (G4C2) repeat expansions in C9orf72, is abundant in C9orf72-related amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) (C9orf72-ALS/FTD). Although the poly-GA aggregates have been identified in C9orf72-ALS/FTD neurons, the effects on UPS (ubiquitin-proteasome system) and autophagy and their exact molecular mechanisms have not been fully elucidated. RESULTS Herein, our in vivo experiments indicate that the mice expressing ploy-GA with 150 repeats instead of 30 repeats exhibit significant aggregates in cells. Mice expressing 150 repeats ploy-GA shows behavioral deficits and activates autophagy in the brain. In vitro findings suggest that the poly-GA aggregates influence proteasomal by directly binding proteasome subunit PSMD2. Subsequently, the poly-GA aggregates activate phosphorylation and ubiquitination of p62 to recruit autophagosomes. Ultimately, the poly-GA aggregates lead to compensatory activation of autophagy. In vivo studies further reveal that rapamycin (autophagy activator) treatment significantly improves the degenerative symptoms and alleviates neuronal injury in mice expressing 150 repeats poly-GA. Meanwhile, rapamycin administration to mice expressing 150 repeats poly-GA reduces neuroinflammation and aggregates in the brain. CONCLUSION In summary, we elucidate the relationship between poly-GA in the proteasome and autophagy: when poly-GA forms complexes with the proteasome, it recruits autophagosomes and affects proteasome function. Our study provides support for further promoting the comprehension of the pathogenesis of C9orf72, which may bring a hint for the exploration of rapamycin for the treatment of ALS/FTD.
Collapse
Affiliation(s)
- Mei Pu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yusi Tai
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Luyang Yuan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Zhang
- Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Huijie Guo
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zongbing Hao
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jing Chen
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| | - Zhouteng Tao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China.
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an, China.
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
42
|
Dominant-negative p53-overexpression in skeletal muscle induces cell death and fiber atrophy in rats. Cell Death Dis 2022; 13:716. [PMID: 35977948 PMCID: PMC9385859 DOI: 10.1038/s41419-022-05160-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 01/21/2023]
Abstract
The tumor suppressor p53 is thought to play a key role in the maintenance of cell size and homeostasis, but relatively little is known about its role in skeletal muscle. Based on its ability to suppress cell growth, we hypothesized that inhibiting the function of wild-type p53 through the overexpression of a dominant-negative p53 mutant (DDp53) could result in muscle fiber hypertrophy. To test this hypothesis, we electroporated adult rat tibialis anterior muscles with DDp53 and collected the tissue three weeks later. We confirmed successful overexpression of DDp53 on a histological and biochemical level and found pronounced changes to muscle architecture, metabolism, and molecular signaling. Muscle mass, fiber cross-sectional area, and fiber diameter significantly decreased with DDp53 overexpression. We found histopathological changes in DDp53 transfected muscle which were accompanied by increased levels of proteins that are associated with membrane damage and repair. In addition, DDp53 decreased oxidative phosphorylation complex I and V protein levels, and despite its negative effects on muscle mass and fiber size, caused an increase in muscle protein synthesis as assessed via the SUnSET technique. Interestingly, the increase in muscle protein synthesis was concomitant with a decrease in phospho-S6K1 (Thr389). Furthermore, the muscle wasting in the DDp53 electroporated leg was accompanied by a decrease in global protein ubiquitination and an increase in proteasome activity. In conclusion, overexpression of a dominant-negative p53 mutant in skeletal muscle results in decreased muscle mass, myofiber size, histological muscle damage, a metabolic phenotype, and perturbed homeostasis between muscle protein synthesis and degradation.
Collapse
|
43
|
Wolska-Washer A, Smolewski P. Targeting Protein Degradation Pathways in Tumors: Focusing on their Role in Hematological Malignancies. Cancers (Basel) 2022; 14:3778. [PMID: 35954440 PMCID: PMC9367439 DOI: 10.3390/cancers14153778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/12/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Cells must maintain their proteome homeostasis by balancing protein synthesis and degradation. This is facilitated by evolutionarily-conserved processes, including the unfolded protein response and the proteasome-based system of protein clearance, autophagy, and chaperone-mediated autophagy. In some hematological malignancies, including acute myeloid leukemia, misfolding or aggregation of the wild-type p53 tumor-suppressor renders cells unable to undergo apoptosis, even with an intact p53 DNA sequence. Moreover, blocking the proteasome pathway triggers lymphoma cell apoptosis. Extensive studies have led to the development of proteasome inhibitors, which have advanced into drugs (such as bortezomib) used in the treatment of certain hematological tumors, including multiple myeloma. New therapeutic options have been studied making use of the so-called proteolysis-targeting chimeras (PROTACs), that bind desired proteins with a linker that connects them to an E3 ubiquitin ligase, resulting in proteasomal-targeted degradation. This review examines the mechanisms of protein degradation in the cells of the hematopoietic system, explains the role of dysfunctional protein degradation in the pathogenesis of hematological malignancies, and discusses the current and future advances of therapies targeting these pathways, based on an extensive search of the articles and conference proceedings from 2005 to April 2022.
Collapse
Affiliation(s)
| | - Piotr Smolewski
- Department of Experimental Hematology, Medical University of Lodz, 93-510 Lodz, Poland;
| |
Collapse
|
44
|
Taylor E, Kim Y, Zhang K, Chau L, Nguyen BC, Rayalam S, Wang X. Antiaging Mechanism of Natural Compounds: Effects on Autophagy and Oxidative Stress. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144396. [PMID: 35889266 PMCID: PMC9322024 DOI: 10.3390/molecules27144396] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 12/22/2022]
Abstract
Aging is a natural biological process that manifests as the progressive loss of function in cells, tissues, and organs. Because mechanisms that are meant to promote cellular longevity tend to decrease in effectiveness with age, it is no surprise that aging presents as a major risk factor for many diseases such as cardiovascular disease, neurodegenerative disorders, cancer, and diabetes. Oxidative stress, an imbalance between the intracellular antioxidant and overproduction of reactive oxygen species, is known to promote the aging process. Autophagy, a major pathway for protein turnover, is considered as one of the hallmarks of aging. Given the progressive physiologic degeneration and increased risk for disease that accompanies aging, many studies have attempted to discover new compounds that may aid in the reversal of the aging process. Here, we summarize the antiaging mechanism of natural or naturally derived synthetic compounds involving oxidative stress and autophagy. These compounds include: 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) derivatives (synthetic triterpenoids derived from naturally occurring oleanolic acid), caffeic acid phenethyl ester (CAPE, the active ingredient in honey bee propolis), xanthohumol (a prenylated flavonoid identified in the hops plant), guggulsterone (a plant steroid found in the resin of the guggul plant), resveratrol (a natural phenol abundantly found in grape), and sulforaphane (a sulfur-containing compound found in cruciferous vegetables).
Collapse
Affiliation(s)
- Elizabeth Taylor
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA;
| | - Yujin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA 30024, USA; (Y.K.); (K.Z.); (L.C.); (B.C.N.); (S.R.)
| | - Kaleb Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA 30024, USA; (Y.K.); (K.Z.); (L.C.); (B.C.N.); (S.R.)
| | - Lenne Chau
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA 30024, USA; (Y.K.); (K.Z.); (L.C.); (B.C.N.); (S.R.)
| | - Bao Chieu Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA 30024, USA; (Y.K.); (K.Z.); (L.C.); (B.C.N.); (S.R.)
| | - Srujana Rayalam
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA 30024, USA; (Y.K.); (K.Z.); (L.C.); (B.C.N.); (S.R.)
| | - Xinyu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine-Georgia Campus, Suwanee, GA 30024, USA; (Y.K.); (K.Z.); (L.C.); (B.C.N.); (S.R.)
- Correspondence:
| |
Collapse
|
45
|
Resveratrol as a modulatory of apoptosis and autophagy in cancer therapy. Clin Transl Oncol 2022; 24:1219-1230. [PMID: 35038152 DOI: 10.1007/s12094-021-02770-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/24/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the leading causes of death, with a heavy socio-economical burden for countries. Despite the great advances that have been made in the treatment of cancer, chemotherapy is still the most common method of treatment. However, many side effects, including hepatotoxicity, renal toxicity, and cardiotoxicity, limit the efficacy of conventional chemotherapy. Over recent years, natural products have attracted attention as therapeutic agents against various diseases, such as cancer. Resveratrol (RES), a natural polyphenol occurring in grapes, nuts, wine, and berries, exhibited potential for preventing and treating various cancer types. RES also ameliorates chemotherapy-induced detrimental effects. Furthermore, RES could modulate apoptosis and autophagy as the main forms of cancer cell deaths by targeting various signaling pathways and up/downregulation of apoptotic and autophagic genes. This review will summarize the anti-cancer effects of RES and focus on the fundamental mechanisms and targets for modulating apoptosis and autophagy by RES.
Collapse
|
46
|
Luong AM, Koestel J, Bhati KK, Batoko H. Cargo receptors and adaptors for selective autophagy in plant cells. FEBS Lett 2022; 596:2104-2132. [PMID: 35638898 DOI: 10.1002/1873-3468.14412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/08/2022] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Abstract
Plant selective (macro)autophagy is a highly regulated process whereby eukaryotic cells spatiotemporally degrade some of their constituents that have become superfluous or harmful. The identification and characterization of the factors determining this selectivity make it possible to integrate selective (macro)autophagy into plant cell physiology and homeostasis. The specific cargo receptors and/or scaffold proteins involved in this pathway are generally not structurally conserved, as are the biochemical mechanisms underlying recognition and integration of a given cargo into the autophagosome in different cell types. This review discusses the few specific cargo receptors described in plant cells to highlight key features of selective autophagy in the plant kingdom and its integration with plant physiology, so as to identify evolutionary convergence and knowledge gaps to be filled by future research.
Collapse
Affiliation(s)
- Ai My Luong
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| | - Jérôme Koestel
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| | - Kaushal Kumar Bhati
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| | - Henri Batoko
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| |
Collapse
|
47
|
SQSTM1, a protective factor of SOD1-linked motor neuron disease, regulates the accumulation and distribution of ubiquitinated protein aggregates in neuron. Neurochem Int 2022; 158:105364. [DOI: 10.1016/j.neuint.2022.105364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/04/2022] [Accepted: 05/26/2022] [Indexed: 11/19/2022]
|
48
|
Promsan S, Thongnak L, Pengrattanachot N, Phengpol N, Sutthasupha P, Lungkaphin A. Agomelatine, a structural analog of melatonin, improves kidney dysfunction through regulating the AMPK/mTOR signaling pathway to promote autophagy in obese rats. Food Chem Toxicol 2022; 165:113190. [DOI: 10.1016/j.fct.2022.113190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 12/20/2022]
|
49
|
Zhou J, Wang F, Jia L, Chai R, Wang H, Wang X, Li J, Wang K, Zhang P, Yang H. 2,4-dichlorophenoxyacetic acid induces ROS activation in NLRP3 inflammatory body-induced autophagy disorder in microglia and the protective effect of Lycium barbarum polysaccharide. ENVIRONMENTAL TOXICOLOGY 2022; 37:1136-1151. [PMID: 35099110 DOI: 10.1002/tox.23471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/26/2021] [Accepted: 01/16/2022] [Indexed: 06/14/2023]
Abstract
The pesticide 2,4-dichlorophenoxyacetic acid (2,4-D) exerts neurotoxic effects; however, its action mechanism remains unclear. Here, we used BV2 cells as a model and divided them into six groups: control group (serum-free medium), lipopolysaccharide (LPS) (1 μg/mL), 2,4-D (1.2 μmol/mL), Lycium barbarum polysaccharide (LBP; 300 μg/mL LBP), LPS (1 μg/mL) + LBP (300 μg/mL), and 2,4-D (1.2 μmol/mL) + LBP (300 μg/mL) with dimethyl sulfoxide as the solvent. Our results showed that 2,4-D treatment decreased superoxide dismutase and glutathione peroxidase activities and increased malondialdehyde content. The percentage of microglial activation (co-expression of ionized calcium-binding adaptor protein-1 + CD68) in the LPS and 2,4-D groups and the levels of tumor necrosis factor alpha, interleukin (IL) 1 beta, IL-6, and IL-18 in the cell supernatant were increased. The protein and mRNA levels of Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein, caspase-1, IL-1β, IL-18, and p62 increased, whereas those of LC3II/I and Beclin-1 decreased in the 2,4-D group. The protein expression and mRNA levels of NLRP3, cleaved caspase-1, IL-1β, IL-18, and p62 decreased significantly, whereas the protein expression and mRNA levels of LC3II/I and Beclin-1 increased in small interfering RNA of NLRP3-treated BV2 cells stimulated with 2,4-D and LPS. In conclusion, 2,4-D enhanced cell migration, promoted oxidative stress, induced excessive release of mitochondrial reactive oxygen species, promoted microglial cell activation, released inflammatory factors, activated NLRP3 inflammasomes, and inhibited autophagy. Meanwhile, LBP reduced inflammation and the release of mitochondrial reactive oxygen species, inhibited NLRP3 inflammasome activation, and regulated autophagy, thereby playing a neuroprotective role.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Faxuan Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Leina Jia
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Ru Chai
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Hengquan Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Xiaolan Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Jiangping Li
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Kai Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Pengju Zhang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Huifang Yang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| |
Collapse
|
50
|
Cross-talk between mutant p53 and p62/SQSTM1 augments cancer cell migration by promoting the degradation of cell adhesion proteins. Proc Natl Acad Sci U S A 2022; 119:e2119644119. [PMID: 35439056 PMCID: PMC9173583 DOI: 10.1073/pnas.2119644119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Missense mutations in the TP53 gene, encoding the p53 tumor suppressor, are very frequent in human cancer. Some of those mutations, particularly the more common (“hotspot”) ones, not only abrogate p53’s tumor suppressor activities but also endow the mutant protein with oncogenic gain of function (GOF). We report that p53R273H, the most common p53 mutant in pancreatic cancer, interacts with the SQSTM1/p62 protein to accelerate the degradation of cell adhesion proteins. This enables pancreatic cancer cells to detach from the epithelial sheet and engage in individualized cell migration, probably augmenting metastatic spread. By providing insights into mechanisms that underpin mutant p53 GOF, this study may suggest ways to interfere with the progression of cancers bearing particular p53 mutants. Missense mutations in the p53 tumor suppressor abound in human cancer. Common (“hotspot”) mutations endow mutant p53 (mutp53) proteins with oncogenic gain of function (GOF), including enhanced cell migration and invasiveness, favoring cancer progression. GOF is usually attributed to transcriptional effects of mutp53. To elucidate transcription-independent effects of mutp53, we characterized the protein interactome of the p53R273H mutant in cells derived from pancreatic ductal adenocarcinoma (PDAC), where p53R273H is the most frequent p53 mutant. We now report that p53R273H, but not the p53R175H hotspot mutant, interacts with SQSTM1/p62 and promotes cancer cell migration and invasion in a p62-dependent manner. Mechanistically, the p53R273H-p62 axis drives the proteasomal degradation of several cell junction–associated proteins, including the gap junction protein Connexin 43, facilitating scattered cell migration. Concordantly, down-regulation of Connexin 43 augments PDAC cell migration, while its forced overexpression blunts the promigratory effect of the p53R273H-p62 axis. These findings define a mechanism of mutp53 GOF.
Collapse
|