1
|
Mallick S, Shormi AS, Jahan H, Alam MS, Begum RA, Sarker RH, Muid KA. Yeast cells experience chronological life span extension under prolonged glucose starvation. Heliyon 2025; 11:e42898. [PMID: 40070955 PMCID: PMC11894305 DOI: 10.1016/j.heliyon.2025.e42898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Budding yeast, Saccharomyces cerevisiae, is an ideal model organism for genetic research due to its similarity in life cycle and cellular structure to higher eukaryotes as well as its ease of cultivation and manipulation in the laboratory. Yeast cells benefit from being cultured in calorie-restricted media, which can be achieved by reducing glucose concentration from 2 % to 0.5 %. Cell metabolism depends on glucose and therefore, affects the physiology of the cell. This study aimed to investigate the effects of long-term glucose starvation on the lifespan of yeast cells by culturing in both standard and glucose-starved conditions. In this investigation yeast cells (BY4743 strain) were cultured in glucose-restricted YPD media (0.5 percent dextrose) to assess lifespan, growth-proliferation, autophagy, apoptosis, mtDNA abundance. The findings revealed that prolonged glucose restriction significantly extended chronological lifespan in yeast (p < 0.05). In order to decipher how starved yeast live chronologically longer, we tested mitochondrial association and found that calorie deprivation lowered the rate of mtDNA spontaneous mutation and increased mtDNA abundance which is a suggestive sign of mitobiogenesis. Furthermore, cells cultured on glucose-restricted media led to more autophagosome formation but less cell death. These results suggested that glucose restriction can enhance lifespan by improving overall cellular conditions. These findings may serve as a foundation for future research in aging, cancer and diabetes.
Collapse
Affiliation(s)
- Setu Mallick
- Genetics and Molecular Biology branch, Department of Zoology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Atia Shanjida Shormi
- Genetics and Molecular Biology branch, Department of Zoology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Hawa Jahan
- Genetics and Molecular Biology branch, Department of Zoology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Mohammad Shamimul Alam
- Genetics and Molecular Biology branch, Department of Zoology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Rowshan Ara Begum
- Genetics and Molecular Biology branch, Department of Zoology, University of Dhaka, Dhaka, 1000, Bangladesh
| | | | - Khandaker Ashfaqul Muid
- Genetics and Molecular Biology branch, Department of Zoology, University of Dhaka, Dhaka, 1000, Bangladesh
| |
Collapse
|
2
|
Garcia-Sanchez JA, Bonnet E, Loubatier C, Doye A, Paillier G, Segui F, Larbret F, Chaintreuil P, Batistic L, Torre C, Deckert M, Polanowska J, Munro P, Boyer L, Visvikis O. Evolutionary conserved regulation of TFEB stability by the E3 ubiquitin ligase WWP2 modulates response to stress in vivo. iScience 2025; 28:111838. [PMID: 39995862 PMCID: PMC11848471 DOI: 10.1016/j.isci.2025.111838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Transcription factor EB (TFEB) is a key transcription factor that orchestrates the cellular response to stress. Dysregulation of TFEB is associated with a range of human diseases, and understanding the regulatory mechanisms of TFEB is crucial for identifying potential drug targets. In this study, we used Caenorhabditis elegans to screen for E3 ubiquitin ligases regulating the activity of TFEB's homolog, HLH-30, upon pathogenic infection. We identified WWP-1 as a regulator of HLH-30-dependent immune response controlling HLH-30 stability to mediate host defense in vivo. We found that HLH-30 interacts with WWP-1, supporting a model of WWP-1 directly regulating HLH-30. Furthermore, we found that WWP-1's human homolog WWP2 binds TFEB, directly induces TFEB ubiquitination and stabilizes TFEB. Finally, we found that WWP2 is required for TFEB-dependent host response in human monocytes-derived macrophages upon infection. Overall, our work has identified an evolutionarily conserved regulation of TFEB by WWP2 and highlighted its role in modulating stress response.
Collapse
Affiliation(s)
| | - Estelle Bonnet
- Université Côte d’Azur, INSERM, C3M, Nice, France
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur, Centre Scientifique de Monaco, Monaco, Monaco
| | | | - Anne Doye
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | - Fabien Segui
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | | | | | - Cédric Torre
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | | | | | | | | |
Collapse
|
3
|
Xu Y, Wang Q, Wang J, Qian C, Wang Y, Lu S, Song L, He Z, Liu W, Wan W. The cGAS-STING pathway activates transcription factor TFEB to stimulate lysosome biogenesis and pathogen clearance. Immunity 2025; 58:309-325.e6. [PMID: 39689715 DOI: 10.1016/j.immuni.2024.11.017] [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: 05/24/2024] [Revised: 09/27/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
Induction of autophagy is an ancient function of the cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway through which autophagic cargoes are delivered to lysosomes for degradation. However, whether lysosome function is also modulated by the cGAS-STING pathway remains unknown. Here, we discovered that the cGAS-STING pathway upregulated lysosomal activity by stimulating lysosome biogenesis independently of the downstream protein kinase TANK-binding kinase 1 (TBK1). STING activation enhanced lysosome biogenesis through inducing the nuclear translocation of transcription factor EB (TFEB) as well as its paralogs transcription factor E3 (TFE3) and microphthalmia-associated transcription factor (MITF). STING-induced lipidation of GABA type A receptor-associated protein (GABARAP), an autophagy-related protein, on STING vesicles was responsible for TFEB activation. Membrane-bound GABARAP sequestered the GTPase-activating protein folliculin (FLCN) and FLCN-interacting protein (FNIP) complex to block its function toward the Rag GTPases Ras-related GTP-binding C and D (RagC and RagD), abolishing mechanistic target of rapamycin (mTOR) complex 1 (mTORC1)-dependent phosphorylation and inactivation of TFEB. Functionally, STING-induced lysosome biogenesis within cells facilitated the clearance of cytoplasmic DNA and invading pathogens. Thus, our findings reveal that induction of lysosome biogenesis is another important function of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Yinfeng Xu
- Laboratory of Basic Biology, Hunan First Normal University, Changsha 410205, Hunan, China.
| | - Qian Wang
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jun Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Chuying Qian
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yusha Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Sheng Lu
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Lijiang Song
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zhengfu He
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Wei Liu
- Department of Metabolic Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, Zhejiang, China.
| | - Wei Wan
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
4
|
Wang Z, Zhang H. Phase-separated Condensates in Autophagosome Formation and Autophagy Regulation. J Mol Biol 2025:168964. [PMID: 39880155 DOI: 10.1016/j.jmb.2025.168964] [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: 11/29/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
Biomacromolecules partition into numerous types of biological condensates or membrane-less organelles via liquid-liquid phase separation (LLPS). Newly formed liquid-like condensates may further undergo phase transition to convert into other material states, such as gel or solid states. Different biological condensates possess distinct material properties to fulfil their physiological functions in diverse cellular pathways and processes. Phase separation and condensates are extensively involved in the autophagy pathway. The autophagosome formation sites in both yeast and multicellular organisms are assembled as phase-separated condensates. TORC1, one of the core regulators of the autophagy-lysosome pathway, is subject to nonconventional regulation by multiple biological condensates. TFEB, the master transcription factor of the autophagy-lysosome pathway, phase separates to assemble liquid-like condensates involved in transcription of autophagic and lysosomal genes. The behaviors and transcriptional activity of TFEB condensates are governed by their material properties, thus suggesting novel autophagy intervention strategies. The phase separation process and the resulting condensates are emerging therapeutic targets for autophagy-related diseases.
Collapse
Affiliation(s)
- Zheng Wang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 PR China; School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006 PR China; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031 PR China.
| | - Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101 PR China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049 PR China.
| |
Collapse
|
5
|
Wang X, Wang L, Zhou Z, Jiang C, Bao Z, Wang Y, Zhang Y, Song L, Zhao Y, Li X, Li Q, Shen Y, Yu Y, Mi W. The ATAC complex represses the transcriptional program of the autophagy-lysosome pathway via its E3 ubiquitin ligase activity. Cell Rep 2024; 43:115033. [PMID: 39643968 DOI: 10.1016/j.celrep.2024.115033] [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: 01/19/2024] [Revised: 07/16/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024] Open
Abstract
The Ada two A-containing (ATAC) complex, containing histone acetyltransferases general control non-derepressible 5 (GCN5) or p300/CBP-associated factor (PCAF), has gained recognition as a prominent transcriptional coactivator. Recent revelations unveiled E3 ligase activity present in both GCN5 and PCAF; however, how the dual enzymatic activities of the ATAC complex orchestrate distinct transcriptional programs and signaling networks remains largely elusive. Our study unveils the function of the ATAC complex as a negative regulator of the autophagy-lysosome pathway's transcriptional program by modulating the stability of transcription factors TFE3 and TFEB. The ATAC complex primarily impacts TFE3/TFEB destabilization through its E3 ligase activity rather than its acetyltransferase function. GCN5/PCAF-mediated ubiquitination prompts the proteasome-dependent degradation of TFE3 and TFEB. Furthermore, inactivation of the ATAC complex amplifies TFE3/TFEB-mediated autophagy-lysosome functions, thereby promoting cell survival during nutrient deprivation. In summary, our findings establish the "ATAC complex-TFE3/TFEB-autophagy-lysosome" axis as an intrinsic regulatory pathway for resisting starvation-induced cell death.
Collapse
Affiliation(s)
- Xiaolu Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Lingling Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhili Zhou
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Chenhao Jiang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Ziyu Bao
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yuexin Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Ying Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lili Song
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yueling Zhao
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xinying Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Qianqian Li
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yujun Shen
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Ying Yu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin 300070, China.
| | - Wenyi Mi
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
6
|
Li Y, Xu Y, Li R, Huang S, Wu Q, Yan J, Jiang Z, Wu X, Li F, Wang Y, Li Y, Fan X, Yuan W. Transcriptomic and Metabolomic Analysis Reveals Multifaceted Impact of Gcn5 Knockdown in Drosophila Development. Metabolites 2024; 14:680. [PMID: 39728461 DOI: 10.3390/metabo14120680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Background: General control nonderepressible 5 (Gcn5) is a lysine acetyltransferase (KAT) that is evolutionarily conserved across eukaryotes, with two homologs (Kat2a and Kat2b) identified in humans and one (Gcn5) in Drosophila. Gcn5 contains a P300/CBP-associated factor (PCAF) domain, a Gcn5-N-acetyltransferase (GNAT) domain, and a Bromodomain, allowing it to regulate gene expression through the acetylation of both histone and non-histone proteins. In Drosophila, Gcn5 is crucial for embryonic development, with maternal Gcn5 supporting early development. However, the functional mechanisms of Gcn5 after the depletion of maternal deposits remain unclear. Methods: Our study employed the Gal4/UAS-RNAi system to achieve whole-body or heart-specific Gcn5 knockdown in Drosophila and selected 96-hour-old surviving larvae for transcriptomic and metabolomic analyses. Results: Omics results revealed that Gcn5 knockdown significantly impacts various metabolic pathways, as well as lysosomes, non-homologous end-joining, Toll and Imd signaling pathways, and circadian rhythms, among others. Furthermore, defects in chitin synthesis may be associated with impaired pupation. Additionally, heart-specific Gcn5 knockdown affected cardiac physiology but appeared to have a potential protective effect against age-related cardiac decline. Conclusions: These findings deepen our understanding of Gcn5's roles in Drosophila development and provide valuable insights for developing Gcn5-targeted therapies, particularly considering its involvement in various human diseases.
Collapse
Affiliation(s)
- Youfeng Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yue Xu
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Ruike Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Sirui Huang
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Qiong Wu
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Jing Yan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zhigang Jiang
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Xiushan Wu
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Fang Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yuequn Wang
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yongqing Li
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Xiongwei Fan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Wuzhou Yuan
- The Laboratory of Heart Development Research, College of Life Science, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
7
|
Keshri S, Vicinanza M, Takla M, Rubinsztein DC. USP7 protects TFEB from proteasome-mediated degradation. Cell Rep 2024; 43:114872. [PMID: 39412987 DOI: 10.1016/j.celrep.2024.114872] [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: 04/25/2024] [Revised: 08/22/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
The transcription factor EB (TFEB) is a master regulator of lysosomal biogenesis and autophagy. We identify a distinct nuclear interactome of TFEB, with ubiquitin-specific protease 7 (USP7) emerging as a key post-translational modulator of TFEB. Genetic depletion and inhibition of USP7 reveal its critical role in preserving TFEB stability within both nuclear and cytoplasmic compartments. Specifically, USP7 is identified as the deubiquitinase responsible for removing the K48-linked polyubiquitination signal from TFEB at lysine residues K116, K264, and K274, thereby preventing its proteasomal degradation. Functional assays demonstrate the involvement of USP7 in preserving TFEB-mediated transcriptional responses to nutrient deprivation while also modulating autophagy flux and lysosome biogenesis. As USP7 is a deubiquitinase that protects TFEB from proteasomal degradation, these findings provide the foundation for therapeutic targeting of the USP7-TFEB axis in conditions characterized by TFEB dysregulation and metabolic abnormalities, particularly in certain cancers.
Collapse
Affiliation(s)
- Swati Keshri
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK; UK Dementia Research Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Mariella Vicinanza
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK; UK Dementia Research Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Michael Takla
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK; UK Dementia Research Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - David C Rubinsztein
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK; UK Dementia Research Institute, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
8
|
Huang Y, Luo G, Peng K, Song Y, Wang Y, Zhang H, Li J, Qiu X, Pu M, Liu X, Peng C, Neculai D, Sun Q, Zhou T, Huang P, Liu W. Lactylation stabilizes TFEB to elevate autophagy and lysosomal activity. J Cell Biol 2024; 223:e202308099. [PMID: 39196068 PMCID: PMC11354204 DOI: 10.1083/jcb.202308099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 02/04/2024] [Accepted: 04/03/2024] [Indexed: 08/29/2024] Open
Abstract
The transcription factor TFEB is a major regulator of lysosomal biogenesis and autophagy. There is growing evidence that posttranslational modifications play a crucial role in regulating TFEB activity. Here, we show that lactate molecules can covalently modify TFEB, leading to its lactylation and stabilization. Mechanically, lactylation at K91 prevents TFEB from interacting with E3 ubiquitin ligase WWP2, thereby inhibiting TFEB ubiquitination and proteasome degradation, resulting in increased TFEB activity and autophagy flux. Using a specific antibody against lactylated K91, enhanced TFEB lactylation was observed in clinical human pancreatic cancer samples. Our results suggest that lactylation is a novel mode of TFEB regulation and that lactylation of TFEB may be associated with high levels of autophagy in rapidly proliferating cells, such as cancer cells.
Collapse
Affiliation(s)
- Yewei Huang
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Gan Luo
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Kesong Peng
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yue Song
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yusha Wang
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hongtao Zhang
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jin Li
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xiangmin Qiu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Maomao Pu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xinchang Liu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Chao Peng
- National Center for Protein Science Shanghai, Institute of Biochemistry and Cell Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dante Neculai
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qiming Sun
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Tianhua Zhou
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Pintong Huang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Lin J, Yuan Y, Huang C, Zi J, Li L, Liu J, Wu X, Li W, Zhao Q, Li Y, Liu Z, Diao A. TFEB agonist clomiphene citrate activates the autophagy-lysosomal pathway and ameliorates Alzheimer's disease symptoms in mice. J Biol Chem 2024; 300:107929. [PMID: 39454957 PMCID: PMC11599454 DOI: 10.1016/j.jbc.2024.107929] [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: 07/19/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Autophagy is a conserved eukaryotic cellular clearance and recycling process through the lysosome-mediated degradation of damaged organelles and protein aggregates to maintain homeostasis. Impairment of the autophagy-lysosomal pathway is implicated in the pathogenesis of Alzheimer's disease (AD). Transcription factor EB (TFEB) is a master regulator of autophagy and lysosomal biogenesis. Therefore, activating TFEB and autophagy provides a novel strategy for AD treatment. We previously described that clomiphene citrate (CC) promotes nuclear translocation of TFEB and increases autophagy and lysosomal biogenesis. In this study, 7- and 3-month-old APP/PS1 mice were treated with TFEB agonist CC and assessed. The behavioral tests were performed using Morris water maze and open field test. Additional changes in amyloid-β pathology, autophagy, and inflammatory response were determined. We found that CC activated TFEB and the autophagy-lysosomal pathway in neuronal cells. Moreover, using mouse model of Alzheimer's disease, CC treatment promoted clearance of amyloid-β plaques and ameliorated cognitive function in both 7- and 3-month-old APP/PS1 mice. The CC-induced activation of TFEB occurs by promoting acetylation of TFEB for nuclear translocation. These findings provide a molecular mechanism for the TFEB-mediated activation of the autophagy-lysosome pathway by CC, which has the potential to be repurposed and applied in the treatment or prevention of AD.
Collapse
Affiliation(s)
- Jieru Lin
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yi Yuan
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Chunhuan Huang
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jiayu Zi
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Lu Li
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jiamiao Liu
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xiaoting Wu
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Wei Li
- School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Qing Zhao
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yuyin Li
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Zhenxing Liu
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| | - Aipo Diao
- School of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
10
|
Colino-Lage H, Guerrero-Gómez D, Gómez-Orte E, González X, Martina JA, Dansen TB, Ayuso C, Askjaer P, Puertollano R, Irazoqui JE, Cabello J, Miranda-Vizuete A. Regulation of Caenorhabditis elegans HLH-30 subcellular localization dynamics: Evidence for a redox-dependent mechanism. Free Radic Biol Med 2024; 223:369-383. [PMID: 39059513 PMCID: PMC11977398 DOI: 10.1016/j.freeradbiomed.2024.07.027] [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: 03/14/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Basic Helix-Loop-Helix (bHLH) transcription factors TFEB/TFE3 and HLH-30 are key regulators of autophagy induction and lysosomal biogenesis in mammals and C. elegans, respectively. While much is known about the regulation of TFEB/TFE3, how HLH-30 subcellular dynamics and transactivation are modulated are yet poorly understood. Thus, elucidating the regulation of C. elegans HLH-30 will provide evolutionary insight into the mechanisms governing the function of bHLH transcription factor family. We report here that HLH-30 is retained in the cytoplasm mainly through its conserved Ser201 residue and that HLH-30 physically interacts with the 14-3-3 protein FTT-2 in this location. The FoxO transcription factor DAF-16 is not required for HLH-30 nuclear translocation upon stress, despite that both proteins partner to form a complex that coordinately regulates several organismal responses. Similar as described for DAF-16, the importin IMB-2 assists HLH-30 nuclear translocation, but constitutive HLH-30 nuclear localization is not sufficient to trigger its distinctive transcriptional response. Furthermore, we identify FTT-2 as the target of diethyl maleate (DEM), a GSH depletor that causes a transient nuclear translocation of HLH-30. Together, our work demonstrates that the regulation of TFEB/TFE3 and HLH-30 family members is evolutionarily conserved and that, in addition to a direct redox regulation through its conserved single cysteine residue, HLH-30 can also be indirectly regulated by a redox-dependent mechanism, probably through FTT-2 oxidation.
Collapse
Affiliation(s)
- Hildegard Colino-Lage
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - David Guerrero-Gómez
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eva Gómez-Orte
- Centro de Investigación Biomédica de la Rioja (CIBIR), Logroño, La Rioja, Spain
| | - Xavier González
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tobias B Dansen
- Center for Molecular Medicine, University Medical Center Utrecht, CG Utrecht, the Netherlands
| | - Cristina Ayuso
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Seville, Spain
| | - Peter Askjaer
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Seville, Spain
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Javier E Irazoqui
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Juan Cabello
- Centro de Investigación Biomédica de la Rioja (CIBIR), Logroño, La Rioja, Spain.
| | - Antonio Miranda-Vizuete
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
11
|
Liao M, Zheng W, Wang Y, Li M, Sun X, Liu N, Yao J, Dong F, Wang Q, Ma Y, Mou J. LINC00887 promotes GCN5-dependent H3K27cr level and CRC metastasis via recruitment of YEATS2 and enhancing ETS1 expression. Cell Death Dis 2024; 15:711. [PMID: 39349460 PMCID: PMC11443008 DOI: 10.1038/s41419-024-07091-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/02/2024]
Abstract
Recent observations have revealed upregulation of H3K27cr in colorectal cancer (CRC) tissues; however, the underlying cause remains elusive. This study aimed to investigate the mechanism of H3K27cr upregulation and its roles in CRC metastasis. Clinically, our findings showed that H3K27cr served as a highly accurate diagnostic marker to distinguish CRC tissues from healthy controls. Elevated levels of LINC00887 and H3K27cr were associated with a poorer prognosis in CRC patients. Functionally, LINC00887 and H3K27cr facilitated the migration and invasion of CRC cells. Mechanistically, LINC00887 interacted with SIRT3 protein. Overexpressed of LINC00887 obstructed the enrichment of SIRT3 within GCN5 promoter, thereby elevating H3K27ac but not H3K27cr level within this region, subsequently activating GCN5 expression. This activation increased the global level of H3K27cr, promoting the enrichment of GCN5, H3K27cr, and YEATS2 within ETS1 promoter, activating ETS1 transcription and ultimately promoting the metastasis of CRC. The in vivo study demonstrated that inhibition of LINC00887 suppressed CRC metastasis, but this inhibitory effect was nullified when mice were treated with NaCr. In conclusion, our results confirmed the diagnostic biomarker potential of H3K27cr in individuals with CRC, and proposed a functional model to elucidate the involvement of LINC00887 in promoting CRC metastasis by elevating H3K27cr level.
Collapse
Affiliation(s)
- Meijian Liao
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Wendan Zheng
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Yifan Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Mengting Li
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Xiaolin Sun
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Nan Liu
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130061, PR China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Jia Yao
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Fuxing Dong
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China
| | - Yu Ma
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, PR China.
| | - Jie Mou
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, PR China.
| |
Collapse
|
12
|
Lu J, Su P, Zhao F, Yu K, Yang X, Lv H, Wang D, Zhang J. The role of TFEB-mediated autophagy-lysosome dysfunction in manganese neurotoxicity. Curr Res Toxicol 2024; 7:100193. [PMID: 39381497 PMCID: PMC11459403 DOI: 10.1016/j.crtox.2024.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
Excessive long-term manganese intake can inflict irreversible damage to the nervous system, with a predominant effect on the substantia nigra-striatum pathway. Through a mouse model simulating manganese exposure, we delved into its implications on the central nervous motor system, uncovering autophagy-lysosome dysfunction as a pivotal factor in manganese-induced neurotoxicity. Our research illuminated the molecular mechanisms behind TFEB's role in manganese-triggered neuronal autophagy dysfunction, offering insights into the cellular and molecular mechanisms of manganese-induced abnormal protein accumulation. This study lays a significant theoretical foundation for future endeavors aimed at safeguarding against manganese neurotoxicity. Furthermore, TFEB emerges as a potential early molecular biomarker for manganese exposure, providing a solid basis for preemptive protection and clinical treatment for populations exposed to manganese.
Collapse
Affiliation(s)
- Jiaqiao Lu
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| | - Peng Su
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| | - Fang Zhao
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| | - Kailun Yu
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| | - Xunbo Yang
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| | - Hui Lv
- Department of Health Service Teaching and Research, Dalian Health Service Training Center of Chinese PLA, Da Lian 116001, China
| | - Diya Wang
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| | - Jianbin Zhang
- Department of Occupational and Environmental Health, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, No.169 Chang Le West Rd., Xi’an, Shaanxi 710032, China
| |
Collapse
|
13
|
Liu H, Xie Z, Gao X, Wei L, Li M, Lin Z, Huang X. Lysosomal dysfunction-derived autophagy impairment of gingival epithelial cells in diabetes-associated periodontitis with altered protein acetylation. Cell Signal 2024; 121:111273. [PMID: 38950874 DOI: 10.1016/j.cellsig.2024.111273] [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: 02/27/2024] [Revised: 06/08/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024]
Abstract
Diabetes-associated periodontitis (DP) presents severe inflammation and resistance to periodontal conventional treatment, presenting a significant challenge in clinical management. In this study, we investigated the underlying mechanism driving the hyperinflammatory response in gingival epithelial cells (GECs) of DP patients. Our findings indicate that lysosomal dysfunction under high glucose conditions leads to the blockage of autophagy flux, exacerbating inflammatory response in GECs. Single-cell RNA sequencing and immunohistochemistry analyses of clinical gingival epithelia revealed dysregulation in the lysosome pathway characterized by reduced levels of lysosome-associated membrane glycoprotein 2 (LAMP2) and V-type proton ATPase 16 kDa proteolipid subunit c (ATP6V0C) in subjects with DP. In vitro stimulation of human gingival epithelial cells (HGECs) with a hyperglycemic microenvironment showed elevated release of proinflammatory cytokines, compromised lysosomal acidity and blocked autophagy. Moreover, HGECs with deficiency in ATP6V0C demonstrated impaired autophagy and heightened inflammatory response, mirroring the effects of high glucose stimulation. Proteomic analysis of acetylation modifications identified altered acetylation levels in 28 autophagy-lysosome pathway-related proteins and 37 sites in HGECs subjected to high glucose stimulation or siATP6V0C. Overall, our finding highlights the pivotal role of lysosome impairment in autophagy obstruction in DP and suggests a potential impact of altered acetylation of relevant proteins on the interplay between lysosome dysfunction and autophagy blockage. These insights may pave the way for the development of effective therapeutic strategies against DP.
Collapse
Affiliation(s)
- Hui Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Zhuo Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Xianling Gao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Linhesheng Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Mengdi Li
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Zhengmei Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China.
| | - Xin Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China.
| |
Collapse
|
14
|
Lv C, Li R, Yang D, Song S, Cheng X, Chen T, Chen L, Xiong Y. Broad-spectrum antiviral effect of MoringaA-loaded exosomes against IAV by mediating the GCN5-TFEB-autolysosome pathway. J Med Virol 2024; 96:e29906. [PMID: 39262090 DOI: 10.1002/jmv.29906] [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: 06/04/2024] [Revised: 08/02/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
Influenza virus-induced viral pneumonia is a major threat to human health, and specific therapeutic agents for viral pneumonia are still lacking. MoringaA (MA) is an anti-influenza virus active compound isolated from Moringa seeds, which can inhibit influenza virus by activating the TFEB-autophagic lysosomal pathway in host cells. In this study, we obtained exosomes from M2-type macrophages and encapsulated and delivered MA (MA-Exos), and we investigated the efficacy of MA-Exos in antiviral and viral pneumonia in vivo and in vitro, respectively. In addition, we provided insights into the mechanism by which MA-Exos regulates TFEB-lysosomal autophagy by RNA sequencing. The MA-Exos showed broad-spectrum inhibition of IAV, and significant promotion of the autophagic lysosomal pathway. Meanwhile, we found that GCN5 gene and protein were significantly down-regulated in IAV-infected cells after MA-Exos intervention, indicating its blocking the acetylation of TFEB by GCN5. In addition, MA-Exos also significantly promoted autophagy in lung tissue cells of mice with viral pneumonia. MA-Exos can inhibit and clear influenza virus by mediating the TFEB-autophagy lysosomal pathway by a mechanism related to the down-regulation of histone acetyltransferase GCN5. Our study provides a strategy for targeting MA-Exos for the treatment of viral pneumonia from both antiviral and virus-induced inflammation inhibition pathways.
Collapse
Affiliation(s)
- Chunmei Lv
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ruidong Li
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dandan Yang
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shunqiang Song
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xu Cheng
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Tingting Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Lei Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yongai Xiong
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
15
|
Jiang J, Ren R, Fang W, Miao J, Wen Z, Wang X, Xu J, Jin H. Lysosomal biogenesis and function in osteoclasts: a comprehensive review. Front Cell Dev Biol 2024; 12:1431566. [PMID: 39170917 PMCID: PMC11335558 DOI: 10.3389/fcell.2024.1431566] [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: 05/12/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Lysosomes serve as catabolic centers and signaling hubs in cells, regulating a multitude of cellular processes such as intracellular environment homeostasis, macromolecule degradation, intracellular vesicle trafficking and autophagy. Alterations in lysosomal level and function are crucial for cellular adaptation to external stimuli, with lysosome dysfunction being implicated in the pathogenesis of numerous diseases. Osteoclasts (OCs), as multinucleated cells responsible for bone resorption and maintaining bone homeostasis, have a complex relationship with lysosomes that is not fully understood. Dysregulated function of OCs can disrupt bone homeostasis leading to the development of various bone disorders. The regulation of OC differentiation and bone resorption for the treatment of bone disease have received considerable attention in recent years, yet the role and regulation of lysosomes in OCs, as well as the potential therapeutic implications of intervening in lysosomal biologic behavior for the treatment of bone diseases, remain relatively understudied. This review aims to elucidate the mechanisms involved in lysosomal biogenesis and to discuss the functions of lysosomes in OCs, specifically in relation to differentiation, bone resorption, and autophagy. Finally, we explore the potential therapeutic implication of targeting lysosomes in the treatment of bone metabolic disorders.
Collapse
Affiliation(s)
- Junchen Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Rufeng Ren
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Weiyuan Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiansen Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zijun Wen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haiming Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Dong PF, Liu TB, Chen K, Li D, Li Y, Lian CY, Wang ZY, Wang L. Cadmium targeting transcription factor EB to inhibit autophagy-lysosome function contributes to acute kidney injury. J Adv Res 2024:S2090-1232(24)00297-2. [PMID: 39033876 DOI: 10.1016/j.jare.2024.07.013] [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: 10/06/2023] [Revised: 01/03/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024] Open
Abstract
INTRODUCTION Environmental and occupational exposure to cadmium (Cd) has been shown to cause acute kidney injury (AKI). Previous studies have demonstrated that autophagy inhibition and lysosomal dysfunction are important mechanisms of Cd-induced AKI. OBJECTIVES Transcription factor EB (TFEB) is a critical transcription regulator that modulates autophagy-lysosome function, but its role in Cd-induced AKI is yet to be elucidated. Thus, in vivo and in vitro studies were conducted to clarify this issue. METHODS AND RESULTS Data firstly showed that reduced TFEB expression and nuclear translocation were evident in Cd-induced AKI models, accompanied by autophagy-lysosome dysfunction. Pharmacological and genetic activation of TFEB improved Cd-induced AKI via alleviating autophagy inhibition and lysosomal dysfunction, whereas Tfeb knockdown further aggravated this phenomenon, suggesting the key role of TFEB in Cd-induced AKI by regulating autophagy. Mechanistically, Cd activated mechanistic target of rapamycin complex 1 (mTORC1) to enhance TFEB phosphorylation and thereby inhibiting TFEB nuclear translocation. Cd also activated chromosome region maintenance 1 (CRM1) to promote TFEB nuclear export. Meanwhile, Cd activated general control non-repressed protein 5 (GCN5) to enhance nuclear TFEB acetylation, resulting in the decreased TFEB transcriptional activity. Moreover, inhibition of CRM1 or GCN5 alleviated Cd-induced AKI by enhancing TFEB activity, respectively. CONCLUSION In summary, these findings reveal that TFEB phosphorylation, nuclear export and acetylation independently suppress TFEB activity to cause Cd-induced AKI via regulating autophagy-lysosome function, suggesting that TFEB activation might be a promising treatment strategy for Cd-induced AKI.
Collapse
Affiliation(s)
- Peng-Fei Dong
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Tian-Bin Liu
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City, 250101, Shandong Province, China
| | - Kai Chen
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City, 250101, Shandong Province, China
| | - Dan Li
- Shandong Medicine Technician College, 999 Fengtian Street, Tai'an City, 271016, Shandong Province, China
| | - Yue Li
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Cai-Yu Lian
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Zhen-Yong Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China
| | - Lin Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271017, Shandong Province, China.
| |
Collapse
|
17
|
Xiang Y, Zhong Y, Lai X, Fang Z, Su G, Lv Y, Tang X, Ouyang L, Gao X, Zheng H, He L, Chen J, Huang J, Lai T. Myeloid-specific Hdac10 deletion protects against LPS-induced acute lung injury via P62 acetylation at lysine 165. Respir Res 2024; 25:263. [PMID: 38956592 PMCID: PMC11221109 DOI: 10.1186/s12931-024-02891-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Aberrant activation of macrophages is associated with pathogenesis of acute lung injury (ALI). However, the potential pathogenesis has not been explored. OBJECTIVES We aimed to identify whether histone deacetylase (HDAC) 10 is involved in lipopolysaccharide (LPS)-exposed ALI and reveal the underlying pathogenesis by which it promotes lung inflammation in LPS-exposed ALI via modifying P62 with deacetylation. METHODS We constructed an ALI mice model stimulated with LPS to determine the positive effect of Hdac10 deficiency. Moreover, we cultured murine alveolar macrophage cell line (MH-S cells) and primary bone marrow-derived macrophages (BMDMs) to explore the pro-inflammatory activity and mechanism of HDAC10 after LPS challenge. RESULTS HDAC10 expression was increased both in mice lung tissues and macrophage cell lines and promoted inflammatory cytokines production exposed to LPS. Hdac10 deficiency inhibited autophagy and inflammatory response after LPS stimulation. In vivo, Hdac10fl/fl-LysMCre mice considerably attenuated lung inflammation and inflammatory cytokines release exposed to LPS. Mechanistically, HDAC10 interacts with P62 and mediates P62 deacetylation at lysine 165 (K165), by which it promotes P62 expression and increases inflammatory cytokines production. Importantly, we identified that Salvianolic acid B (SAB), an HDAC10 inhibitor, reduces lung inflammatory response in LPS-stimulated ALI. CONCLUSION These results uncover a previously unknown role for HDAC10 in regulating P62 deacetylation and aggravating lung inflammation in LPS-induced ALI, implicating that targeting HDAC10 is an effective therapy for LPS-exposed ALI.
Collapse
Affiliation(s)
- Yuanyuan Xiang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Yu Zhong
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xianwen Lai
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Zhenfu Fang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Guomei Su
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Yingying Lv
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xiantong Tang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Lihuan Ouyang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xiao Gao
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongying Zheng
- The Department of Pulmonary and Critical Care Medicine, The Third People' s Hospital of Xining City, Qinghai, 810005, China
| | - Lilin He
- The Department of Pulmonary and Critical Care Medicine, The Third People' s Hospital of Xining City, Qinghai, 810005, China
| | - Jialong Chen
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523121, China.
| | - Jiewen Huang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China.
- Dongguan Key Laboratory of Immune Inflammation and Metabolism, Dongguan, 523121, China.
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China.
- Dongguan Key Laboratory of Immune Inflammation and Metabolism, Dongguan, 523121, China.
| |
Collapse
|
18
|
Huang L, Guo H. Acetylation modification in the regulation of macroautophagy. ADVANCED BIOTECHNOLOGY 2024; 2:19. [PMID: 39883319 PMCID: PMC11740868 DOI: 10.1007/s44307-024-00027-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 01/31/2025]
Abstract
Macroautophagy, commonly referred to as autophagy, is an evolutionarily conserved cellular process that plays a crucial role in maintaining cellular homeostasis. It orchestrates the delivery of dysfunctional or surplus cellular materials to the vacuole or lysosome for degradation and recycling, particularly during adverse conditions. Over the past few decades, research has unveiled intricate regulatory mechanisms governing autophagy through various post-translational modifications (PTMs). Among these PTMs, acetylation modification has emerged as a focal point in yeast and animal studies. It plays a pivotal role in autophagy by directly targeting core components within the central machinery of autophagy, including autophagy initiation, nucleation, phagophore expansion, and autophagosome maturation. Additionally, acetylation modulates autophagy at the transcriptional level by modifying histones and transcription factors. Despite its well-established significance in yeast and mammals, the role of acetylation in plant autophagy remains largely unexplored, and the precise regulatory mechanisms remain enigmatic. In this comprehensive review, we summarize the current understanding of the function and underlying mechanisms of acetylation in regulating autophagy across yeast, mammals, and plants. We particularly highlight recent advances in deciphering the impact of acetylation on plant autophagy. These insights not only provide valuable guidance but also inspire further scientific inquiries into the intricate role of acetylation in plant autophagy.
Collapse
Affiliation(s)
- Li Huang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Hongwei Guo
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
19
|
Han Y, Zhao H, Li G, Jia J, Guo H, Tan J, Sun X, Li S, Ran Q, Bai C, Gu Y, Li Z, Guan H, Gao S, Zhou PK. GCN5 mediates DNA-PKcs crotonylation for DNA double-strand break repair and determining cancer radiosensitivity. Br J Cancer 2024; 130:1621-1634. [PMID: 38575732 PMCID: PMC11091118 DOI: 10.1038/s41416-024-02636-4] [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: 12/12/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND DNA double-strand break (DSB) induction and repair are important events for determining cell survival and the outcome of cancer radiotherapy. The DNA-dependent protein kinase (DNA-PK) complex functions at the apex of DSBs repair, and its assembly and activity are strictly regulated by post-translation modifications (PTMs)-associated interactions. However, the PTMs of the catalytic subunit DNA-PKcs and how they affect DNA-PKcs's functions are not fully understood. METHODS Mass spectrometry analyses were performed to identify the crotonylation sites of DNA-PKcs in response to γ-ray irradiation. Co-immunoprecipitation (Co-IP), western blotting, in vitro crotonylation assays, laser microirradiation assays, in vitro DNA binding assays, in vitro DNA-PK assembly assays and IF assays were employed to confirm the crotonylation, identify the crotonylase and decrotonylase, and elucidate how crotonylation regulates the activity and function of DNA-PKcs. Subcutaneous xenografts of human HeLa GCN5 WT or HeLa GCN5 siRNA cells in BALB/c nude mice were generated and utilized to assess tumor proliferation in vivo after radiotherapy. RESULTS Here, we reveal that K525 is an important site of DNA-PKcs for crotonylation, and whose level is sharply increased by irradiation. The histone acetyltransferase GCN5 functions as the crotonylase for K525-Kcr, while HDAC3 serves as its dedicated decrotonylase. K525 crotonylation enhances DNA binding activity of DNA-PKcs, and facilitates assembly of the DNA-PK complex. Furthermore, GCN5-mediated K525 crotonylation is indispensable for DNA-PKcs autophosphorylation and the repair of double-strand breaks in the NHEJ pathway. GCN5 suppression significantly sensitizes xenograft tumors of mice to radiotherapy. CONCLUSIONS Our study defines K525 crotonylation of DNA-PKcs is important for the DNA-PK complex assembly and DSBs repair activity via NHEJ pathway. Targeting GCN5-mediated K525 Kcr of DNA-PKcs may be a promising therapeutic strategy for improving the outcome of cancer radiotherapy.
Collapse
Affiliation(s)
- Yang Han
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hongling Zhao
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Gang Li
- School of Public Health, Institute for Environmental Medicine and Radiation Hygiene, University of South China, Hengyang, China
- Department of Hospital Infection Control, Shenzhen Luohu Peoples Hospital, Shenzhen, China
| | - Jin Jia
- School of Medicine, University of South China, Hengyang, China
| | - Hejiang Guo
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jinpeng Tan
- School of Medicine, University of South China, Hengyang, China
| | - Xingyao Sun
- School of Medicine, University of South China, Hengyang, China
| | - Saiyu Li
- School of life Sciences, Hebei University, Baoding, China
| | - Qian Ran
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Chenjun Bai
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yongqing Gu
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - ZhongJun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China.
| | - Hua Guan
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Shanshan Gao
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China.
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China.
- School of Public Health, Institute for Environmental Medicine and Radiation Hygiene, University of South China, Hengyang, China.
| |
Collapse
|
20
|
Xu X, Wang J, Xia Y, Yin Y, Zhu T, Chen F, Hai C. Autophagy, a double-edged sword for oral tissue regeneration. J Adv Res 2024; 59:141-159. [PMID: 37356803 PMCID: PMC11081970 DOI: 10.1016/j.jare.2023.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/10/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Oral health is of fundamental importance to maintain systemic health in humans. Stem cell-based oral tissue regeneration is a promising strategy to achieve the recovery of impaired oral tissue. As a highly conserved process of lysosomal degradation, autophagy induction regulates stem cell function physiologically and pathologically. Autophagy activation can serve as a cytoprotective mechanism in stressful environments, while insufficient or over-activation may also lead to cell function dysregulation and cell death. AIM OF REVIEW This review focuses on the effects of autophagy on stem cell function and oral tissue regeneration, with particular emphasis on diverse roles of autophagy in different oral tissues, including periodontal tissue, bone tissue, dentin pulp tissue, oral mucosa, salivary gland, maxillofacial muscle, temporomandibular joint, etc. Additionally, this review introduces the molecular mechanisms involved in autophagy during the regeneration of different parts of oral tissue, and how autophagy can be regulated by small molecule drugs, biomaterials, exosomes/RNAs or other specific treatments. Finally, this review discusses new perspectives for autophagy manipulation and oral tissue regeneration. KEY SCIENTIFIC CONCEPTS OF REVIEW Overall, this review emphasizes the contribution of autophagy to oral tissue regeneration and highlights the possible approaches for regulating autophagy to promote the regeneration of human oral tissue.
Collapse
Affiliation(s)
- Xinyue Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Jia Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Yunlong Xia
- Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Tianxiao Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Faming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Chunxu Hai
- Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China.
| |
Collapse
|
21
|
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
|
22
|
Li X, Wu Y, Wang P, Li Y, Gu J, Zhang Y, Yan S, Hu P. LncRNA XXYLT1-AS2 promotes tumor progression via autophagy inhibition through ubiquitinated degradation of TFEB in hepatocellular carcinoma. Clin Transl Oncol 2024; 26:698-708. [PMID: 37540409 DOI: 10.1007/s12094-023-03294-3] [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: 06/08/2023] [Accepted: 07/23/2023] [Indexed: 08/05/2023]
Abstract
PURPOSE There is compelling evidence that long-stranded non-coding RNAs (lncRNAs) play an important role in the progression of hepatocellular carcinoma (HCC). The aim of this study was to investigate the role of lncRNA XXYLT1 antisense-2 (XXYLT1-AS2) in HCC progression. METHODS Real-time PCR was used to assess the levels of XXYLT1-AS2 in plasma from HCC and normal patients. Cell proliferation, apoptosis, migration, and invasion were monitored, and tumor xenografts were established to investigate the biological functions of XXYLT1-AS2 by gain-of-function and loss-of-function studies in vitro and in vivo, the expression of autophagy biomarkers and transcriptional factor EB (TFEB) was examined by immunoprecipitation, ubiquitination assays, and western blotting. Autophagy inhibitor, 3-methyladenine (3MA), and proteasome inhibitor, MG132, were used to verify the role of autophagy in HCC progression and the effect of XXYLT1-AS2 on TFEB ubiquitination, respectively. RESULTS In this study, we identified that lncRNA XXYLT1-AS2 is highly expressed in HCC plasma and promotes tumor growth in vivo. In functional studies, it was found that silent expression of XXYLT1-AS2 inhibited HCC proliferation, migration, invasion, and activated autophagy of HCC cells, which were attenuated by autophagy inhibitor, 3MA. Mechanistically, XXYLT1-AS2 decreased the protein level of TFEB through promoting its degradation by ubiquitin proteasome pathway. CONCLUSION XXYLT1-AS2 plays an oncogenic role in HCC progression through inhibition of autophagy via promoting the degradation of TFEB, and thus could be a novel target for HCC treatment.
Collapse
Affiliation(s)
- Xuejie Li
- Department of Laboratory Medicine, Jinzhou Medical University Graduate Training Base, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yuqin Wu
- Central Operating Room, Taihe Hospital, Shiyan, 442000, Hubei, People's Republic of China
| | - Pingfeng Wang
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, No. 32, South Renmin Road, Shiyan City, 442000, Hubei, People's Republic of China
| | - Ying Li
- Blood Transfusion Department, Taihe Hospital, Shiyan, 442000, Hubei, People's Republic of China
| | - Jiangxue Gu
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yuan Zhang
- Biomedical Engineering College, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Shirong Yan
- Department of Laboratory Medicine, Jinzhou Medical University Graduate Training Base, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, No. 30, South Renmin Road, Shiyan City, 442000, Hubei, People's Republic of China.
| | - Pei Hu
- Department of Laboratory Medicine, Jinzhou Medical University Graduate Training Base, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, No. 32, South Renmin Road, Shiyan City, 442000, Hubei, People's Republic of China.
| |
Collapse
|
23
|
Settembre C, Perera RM. Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat Rev Mol Cell Biol 2024; 25:223-245. [PMID: 38001393 DOI: 10.1038/s41580-023-00676-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/26/2023]
Abstract
Every cell must satisfy basic requirements for nutrient sensing, utilization and recycling through macromolecular breakdown to coordinate programmes for growth, repair and stress adaptation. The lysosome orchestrates these key functions through the synchronised interplay between hydrolytic enzymes, nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles and regulation of specific gene expression programmes. In this Review, we discuss recent findings on lysosome-dependent signalling pathways, focusing on how the lysosome senses nutrient availability through its physical and functional association with mechanistic target of rapamycin complex 1 (mTORC1) and how, in response, the microphthalmia/transcription factor E (MiT/TFE) transcription factors exert feedback regulation on lysosome biogenesis. We also highlight the emerging interactions of lysosomes with other organelles, which contribute to cellular homeostasis. Lastly, we discuss how lysosome dysfunction contributes to diverse disease pathologies and how inherited mutations that compromise lysosomal hydrolysis, transport or signalling components lead to multi-organ disorders with severe metabolic and neurological impact. A deeper comprehension of lysosomal composition and function, at both the cellular and organismal level, may uncover fundamental insights into human physiology and disease.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Rushika M Perera
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Chauhan N, Patro BS. Emerging roles of lysosome homeostasis (repair, lysophagy and biogenesis) in cancer progression and therapy. Cancer Lett 2024; 584:216599. [PMID: 38135207 DOI: 10.1016/j.canlet.2023.216599] [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: 09/28/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
In the era of personalized therapy, precise targeting of subcellular organelles holds great promise for cancer modality. Taking into consideration that lysosome represents the intersection site in numerous endosomal trafficking pathways and their modulation in cancer growth, progression, and resistance against cancer therapies, the lysosome is proposed as an attractive therapeutic target for cancer treatment. Based on the recent advances, the current review provides a comprehensive understanding of molecular mechanisms of lysosome homeostasis under 3R responses: Repair, Removal (lysophagy) and Regeneration of lysosomes. These arms of 3R responses have distinct role in lysosome homeostasis although their interdependency along with switching between the pathways still remain elusive. Recent advances underpinning the crucial role of (1) ESCRT complex dependent/independent repair of lysosome, (2) various Galectins-based sensing and ubiquitination in lysophagy and (3) TFEB/TFE proteins in lysosome regeneration/biogenesis of lysosome are outlined. Later, we also emphasised how these recent advancements may aid in development of phytochemicals and pharmacological agents for targeting lysosomes for efficient cancer therapy. Some of these lysosome targeting agents, which are now at various stages of clinical trials and patents, are also highlighted in this review.
Collapse
Affiliation(s)
- Nitish Chauhan
- Bio-Organic Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, Maharashtra, 400094, India
| | - Birija Sankar Patro
- Bio-Organic Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, Maharashtra, 400094, India.
| |
Collapse
|
25
|
Chen H, Gong S, Zhang H, Chen Y, Liu Y, Hao J, Liu H, Li X. From the regulatory mechanism of TFEB to its therapeutic implications. Cell Death Discov 2024; 10:84. [PMID: 38365838 PMCID: PMC10873368 DOI: 10.1038/s41420-024-01850-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/18/2024] Open
Abstract
Transcription factor EB (TFEB), known as a major transcriptional regulator of the autophagy-lysosomal pathway, regulates target gene expression by binding to coordinated lysosomal expression and regulation (CLEAR) elements. TFEB are regulated by multiple links, such as transcriptional regulation, post-transcriptional regulation, translational-level regulation, post-translational modification (PTM), and nuclear competitive regulation. Targeted regulation of TFEB has been victoriously used as a treatment strategy in several disease models such as ischemic injury, lysosomal storage disorders (LSDs), cancer, metabolic disorders, neurodegenerative diseases, and inflammation. In this review, we aimed to elucidate the regulatory mechanism of TFEB and its applications in several disease models by targeting the regulation of TFEB as a treatment strategy.
Collapse
Affiliation(s)
- Huixia Chen
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Siqiao Gong
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongyong Zhang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhan-jiang Central Hospital, Zhanjiang, 524001, China
| | - Yongming Chen
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yonghan Liu
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Junfeng Hao
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Huafeng Liu
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Xiaoyu Li
- Institute of Nephrology, and Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
26
|
Sipos F, Műzes G. Sirtuins Affect Cancer Stem Cells via Epigenetic Regulation of Autophagy. Biomedicines 2024; 12:386. [PMID: 38397988 PMCID: PMC10886574 DOI: 10.3390/biomedicines12020386] [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: 12/27/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Sirtuins (SIRTs) are stress-responsive proteins that regulate several post-translational modifications, partly by acetylation, deacetylation, and affecting DNA methylation. As a result, they significantly regulate several cellular processes. In essence, they prolong lifespan and control the occurrence of spontaneous tumor growth. Members of the SIRT family have the ability to govern embryonic, hematopoietic, and other adult stem cells in certain tissues and cell types in distinct ways. Likewise, they can have both pro-tumor and anti-tumor effects on cancer stem cells, contingent upon the specific tissue from which they originate. The impact of autophagy on cancer stem cells, which varies depending on the specific circumstances, is a very intricate phenomenon that has significant significance for clinical and therapeutic purposes. SIRTs exert an impact on the autophagy process, whereas autophagy reciprocally affects the activity of certain SIRTs. The mechanism behind this connection in cancer stem cells remains poorly understood. This review presents the latest findings that position SIRTs at the point where cancer cells and autophagy interact. Our objective is to highlight the various roles of distinct SIRTs in cancer stem cell-related functions through autophagy. This would demonstrate their significance in the genesis and recurrence of cancer and offer a more precise understanding of their treatment possibilities in relation to autophagy.
Collapse
Affiliation(s)
- Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary;
| | | |
Collapse
|
27
|
Shao J, Lang Y, Ding M, Yin X, Cui L. Transcription Factor EB: A Promising Therapeutic Target for Ischemic Stroke. Curr Neuropharmacol 2024; 22:170-190. [PMID: 37491856 PMCID: PMC10788889 DOI: 10.2174/1570159x21666230724095558] [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: 05/08/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 07/27/2023] Open
Abstract
Transcription factor EB (TFEB) is an important endogenous defensive protein that responds to ischemic stimuli. Acute ischemic stroke is a growing concern due to its high morbidity and mortality. Most survivors suffer from disabilities such as numbness or weakness in an arm or leg, facial droop, difficulty speaking or understanding speech, confusion, impaired balance or coordination, or loss of vision. Although TFEB plays a neuroprotective role, its potential effect on ischemic stroke remains unclear. This article describes the basic structure, regulation of transcriptional activity, and biological roles of TFEB relevant to ischemic stroke. Additionally, we explore the effects of TFEB on the various pathological processes underlying ischemic stroke and current therapeutic approaches. The information compiled here may inform clinical and basic studies on TFEB, which may be an effective therapeutic drug target for ischemic stroke.
Collapse
Affiliation(s)
- Jie Shao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Manqiu Ding
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
28
|
Lee YY, Ha J, Kim YS, Ramani S, Sung S, Gil ES, Choo OS, Jang JH, Choung YH. Abnormal Cholesterol Metabolism and Lysosomal Dysfunction Induce Age-Related Hearing Loss by Inhibiting mTORC1-TFEB-Dependent Autophagy. Int J Mol Sci 2023; 24:17513. [PMID: 38139347 PMCID: PMC10743727 DOI: 10.3390/ijms242417513] [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] [Received: 11/16/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Cholesterol is a risk factor for age-related hearing loss (ARHL). However, the effect of cholesterol on the organ of Corti during the onset of ARHL is unclear. We established a mouse model for the ARHL group (24 months, n = 12) and a young group (6 months, n = 12). Auditory thresholds were measured in both groups using auditory brainstem response (ABR) at frequencies of 8, 16, and 32 kHz. Subsequently, mice were sacrificed and subjected to histological analyses, including transmission electron microscopy (TEM), H&E, Sudan Black B (SBB), and Filipin staining, as well as biochemical assays such as IHC, enzymatic analysis, and immunoblotting. Additionally, mRNA extracted from both young and aged cochlea underwent RNA sequencing. To identify the mechanism, in vitro studies utilizing HEI-OC1 cells were also performed. RNA sequencing showed a positive correlation with increased expression of genes related to metabolic diseases, cholesterol homeostasis, and target of rapamycin complex 1 (mTORC1) signaling in the ARHL group as compared to the younger group. In addition, ARHL tissues exhibited increased cholesterol and lipofuscin aggregates in the organ of Corti, lateral walls, and spiral ganglion neurons. Autophagic flux was inhibited by the accumulation of damaged lysosomes and autolysosomes. Subsequently, we observed a decrease in the level of transcription factor EB (TFEB) protein, which regulates lysosomal biosynthesis and autophagy, together with increased mTORC1 activity in ARHL tissues. These changes in TFEB and mTORC1 expression were observed in a cholesterol-dependent manner. Treatment of ARHL mice with atorvastatin, a cholesterol synthesis inhibitor, delayed hearing loss by reducing the cholesterol level and maintaining lysosomal function and autophagy by inhibiting mTORC1 and activating TFEB. The above findings were confirmed using stress-induced premature senescent House Ear Institute organ of Corti 1 (HEI-OC1) cells. The findings implicate cholesterol in the pathogenesis of ARHL. We propose that atorvastatin could prevent ARHL by maintaining lysosomal function and autophagy by inhibiting mTORC1 and activating TFEB during the aging process.
Collapse
Affiliation(s)
- Yun Yeong Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
| | - Jungho Ha
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
- Department of Medical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Young Sun Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
| | - Sivasubramanian Ramani
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
| | - Siung Sung
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
- Department of Medical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Eun Sol Gil
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
- Department of Medical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Oak-Sung Choo
- Department of Otorhinolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Republic of Korea;
| | - Jeong Hun Jang
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
| | - Yun-Hoon Choung
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.Y.L.); (J.H.); (Y.S.K.); (S.R.); (S.S.); (J.H.J.)
- Department of Medical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
29
|
Chowdhury SG, Karmakar P. Revealing the role of epigenetic and post-translational modulations of autophagy proteins in the regulation of autophagy and cancer: a therapeutic approach. Mol Biol Rep 2023; 51:3. [PMID: 38063905 DOI: 10.1007/s11033-023-08961-w] [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: 08/17/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023]
Abstract
Autophagy is a process that is characterized by the destruction of redundant components and the removal of dysfunctional ones to maintain cellular homeostasis. Autophagy dysregulation has been linked to various illnesses, such as neurodegenerative disorders and cancer. The precise transcription of the genes involved in autophagy is regulated by a network of epigenetic factors. This includes histone modifications and histone-modifying enzymes. Epigenetics is a broad category of heritable, reversible changes in gene expression that do not include changes to DNA sequences, such as chromatin remodeling, histone modifications, and DNA methylation. In addition to affecting the genes that are involved in autophagy, the epigenetic machinery can also alter the signals that control this process. In cancer, autophagy plays a dual role by preventing the development of tumors on one hand and this process may suppress tumor progression. This may be the control of an oncogene that prevents autophagy while, conversely, tumor suppression may promote it. The development of new therapeutic strategies for autophagy-related disorders could be initiated by gaining a deeper understanding of its intricate regulatory framework. There is evidence showing that certain machineries and regulators of autophagy are affected by post-translational and epigenetic modifications, which can lead to alterations in the levels of autophagy and these changes can then trigger disease or affect the therapeutic efficacy of drugs. The goal of this review is to identify the regulatory pathways associated with post-translational and epigenetic modifications of different proteins in autophagy which may be the therapeutic targets shortly.
Collapse
Affiliation(s)
| | - Parimal Karmakar
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
30
|
Wu W, Lin L, Zhao Y, Li H, Zhang R. Protein modification regulated autophagy in Bombyx mori and Drosophila melanogaster. Front Physiol 2023; 14:1281555. [PMID: 38028759 PMCID: PMC10665574 DOI: 10.3389/fphys.2023.1281555] [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: 08/22/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Post-translational modifications refer to the chemical alterations of proteins following their biosynthesis, leading to changes in protein properties. These modifications, which encompass acetylation, phosphorylation, methylation, SUMOylation, ubiquitination, and others, are pivotal in a myriad of cellular functions. Macroautophagy, also known as autophagy, is a major degradation of intracellular components to cope with stress conditions and strictly regulated by nutrient depletion, insulin signaling, and energy production in mammals. Intriguingly, in insects, 20-hydroxyecdysone signaling predominantly stimulates the expression of most autophagy-related genes while concurrently inhibiting mTOR activity, thereby initiating autophagy. In this review, we will outline post-translational modification-regulated autophagy in insects, including Bombyx mori and Drosophila melanogaster, in brief. A more profound understanding of the biological significance of post-translational modifications in autophagy machinery not only unveils novel opportunities for autophagy intervention strategies but also illuminates their potential roles in development, cell differentiation, and the process of learning and memory processes in both insects and mammals.
Collapse
Affiliation(s)
- Wenmei Wu
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Luobin Lin
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yuntao Zhao
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Huaqin Li
- Guangzhou Xinhua University, Guangzhou, Guangdong, China
| | - Rongxin Zhang
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Takla M, Keshri S, Rubinsztein DC. The post-translational regulation of transcription factor EB (TFEB) in health and disease. EMBO Rep 2023; 24:e57574. [PMID: 37728021 PMCID: PMC10626434 DOI: 10.15252/embr.202357574] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023] Open
Abstract
Transcription factor EB (TFEB) is a basic helix-loop-helix leucine zipper transcription factor that acts as a master regulator of lysosomal biogenesis, lysosomal exocytosis, and macro-autophagy. TFEB contributes to a wide range of physiological functions, including mitochondrial biogenesis and innate and adaptive immunity. As such, TFEB is an essential component of cellular adaptation to stressors, ranging from nutrient deprivation to pathogenic invasion. The activity of TFEB depends on its subcellular localisation, turnover, and DNA-binding capacity, all of which are regulated at the post-translational level. Pathological states are characterised by a specific set of stressors, which elicit post-translational modifications that promote gain or loss of TFEB function in the affected tissue. In turn, the resulting increase or decrease in survival of the tissue in which TFEB is more or less active, respectively, may either benefit or harm the organism as a whole. In this way, the post-translational modifications of TFEB account for its otherwise paradoxical protective and deleterious effects on organismal fitness in diseases ranging from neurodegeneration to cancer. In this review, we describe how the intracellular environment characteristic of different diseases alters the post-translational modification profile of TFEB, enabling cellular adaptation to a particular pathological state.
Collapse
Affiliation(s)
- Michael Takla
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| | - Swati Keshri
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
- UK Dementia Research Institute, Cambridge Institute for Medical Research (CIMR)University of CambridgeCambridgeUK
| |
Collapse
|
32
|
Liao R, Wu Y, Qin L, Jiang Z, Gou S, Zhou L, Hong Q, Li Y, Shi J, Yao Y, Lai L, Li Y, Liu P, Thiery JP, Qin D, Graf T, Liu X, Li P. BCL11B and the NuRD complex cooperatively guard T-cell fate and inhibit OPA1-mediated mitochondrial fusion in T cells. EMBO J 2023; 42:e113448. [PMID: 37737560 PMCID: PMC10620766 DOI: 10.15252/embj.2023113448] [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: 01/05/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
The nucleosome remodeling and histone deacetylase (NuRD) complex physically associates with BCL11B to regulate murine T-cell development. However, the function of NuRD complex in mature T cells remains unclear. Here, we characterize the fate and metabolism of human T cells in which key subunits of the NuRD complex or BCL11B are ablated. BCL11B and the NuRD complex bind to each other and repress natural killer (NK)-cell fate in T cells. In addition, T cells upregulate the NK cell-associated receptors and transcription factors, lyse NK-cell targets, and are reprogrammed into NK-like cells (ITNKs) upon deletion of MTA2, MBD2, CHD4, or BCL11B. ITNKs increase OPA1 expression and exhibit characteristically elongated mitochondria with augmented oxidative phosphorylation (OXPHOS) activity. OPA1-mediated elevated OXPHOS enhances cellular acetyl-CoA levels, thereby promoting the reprogramming efficiency and antitumor effects of ITNKs via regulating H3K27 acetylation at specific targets. In conclusion, our findings demonstrate that the NuRD complex and BCL11B cooperatively maintain T-cell fate directly by repressing NK cell-associated transcription and indirectly through a metabolic-epigenetic axis, providing strategies to improve the reprogramming efficiency and antitumor effects of ITNKs.
Collapse
Affiliation(s)
- Rui Liao
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Yi Wu
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Le Qin
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Zhiwu Jiang
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Shixue Gou
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Linfu Zhou
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Qilan Hong
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
- Centre for Genomic RegulationThe Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Yao Li
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Jingxuan Shi
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Yao Yao
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Liangxue Lai
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
| | - Yangqiu Li
- Institute of HematologyMedical College, Jinan UniversityGuangzhouChina
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | | | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Thomas Graf
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
- Centre for Genomic RegulationThe Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Xingguo Liu
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & InnovationChinese Academy of SciencesHong Kong SARChina
| | - Peng Li
- China‐New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH‐HKU Guangdong‐Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH‐CUHK Joint Research Laboratory on Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesGuangzhouChina
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education InstitutesThe Fifth Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & InnovationChinese Academy of SciencesHong Kong SARChina
- Department of SurgeryThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
33
|
Li G, Li Y, He C, Wei Y, Cai K, Lu Q, Liu X, Zhu Y, Xu K. The promoting effects of pyriproxyfen on autophagy and apoptosis in silk glands of non-target insect silkworm, Bombyx mori. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 196:105586. [PMID: 37945223 DOI: 10.1016/j.pestbp.2023.105586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/12/2023] [Accepted: 08/23/2023] [Indexed: 11/12/2023]
Abstract
Pyriproxyfen is a juvenile hormone analogue. The physiological effects of its low-concentration drift during the process of controlling agricultural and forestry pests on non-target organisms in the ecological environment are unpredictable, especially the effects on organs that play a key role in biological function are worthy of attention. The silk gland is an important organ for silk-secreting insects. Herein, we studied the effects of trace pyriproxyfen on autophagy and apoptosis of the silk gland in the lepidopteran model insect, Bombyx mori (silkworm). After treating fifth instar silkworm larvae with pyriproxyfen for 24 h, we found significant shrinkage, vacuolization, and fragmentation in the posterior silk gland (PSG). In addition, the results of autophagy-related genes of ATG8 and TUNEL assay also demonstrated that autophagy and apoptosis in the PSG of the silkworm was induced by pyriproxyfen. RNA-Seq results showed that pyriproxyfen treatment resulted in the activation of juvenile hormone signaling pathway genes and inhibition of 20-hydroxyecdysone (20E) signaling pathway genes. Among the 1808 significantly differentially expressed genes, 796 were upregulated and 1012 were downregulated. Among them, 30 genes were identified for autophagy-related signaling pathways, such as NOD-like receptor signaling pathway and mTOR signaling pathway, and 30 genes were identified for apoptosis-related signaling pathways, such as P53 signaling pathway and TNF signaling pathway. Further qRT-PCR and in vitro gland culture studies showed that the autophagy-related genes Atg5, Atg6, Atg12, Atg16 and the apoptosis-related genes Aif, Dronc, Dredd, and Caspase1 were responsive to the treatment of pyriproxyfen, with transcription levels up-regulated from 24 to 72 h. In addition, ATG5, ATG6, and Dronc genes had a more direct response to pyriproxyfen treatment. These results suggested that pyriproxyfen treatment could disrupt the hormone regulation in silkworms, promoting autophagy and apoptosis in the PSG. This study provides more evidence for the research on the damage of juvenile hormone analogues to non-target organisms or organs in the environment, and provides reference information for the scientific and rational use of juvenile hormone pesticides.
Collapse
Affiliation(s)
- Guoli Li
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Yizhe Li
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Chunhui He
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Yuting Wei
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Kunpei Cai
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Qingyu Lu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xuebin Liu
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Yizhou Zhu
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China
| | - Kaizun Xu
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China; Guangxi Key Laboratory for Agro-Environment and Agric-Product Safety, National Demonstration Center for Experimental Plant Science Education, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, PR China; Sericulture Institute of Guangxi University, Nanning, Guangxi 530004, PR China.
| |
Collapse
|
34
|
Venkatasubramani AV, Ichinose T, Kanno M, Forne I, Tanimoto H, Peleg S, Imhof A. The fruit fly acetyltransferase chameau promotes starvation resilience at the expense of longevity. EMBO Rep 2023; 24:e57023. [PMID: 37724628 PMCID: PMC10561354 DOI: 10.15252/embr.202357023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023] Open
Abstract
Proteins involved in cellular metabolism and molecular regulation can extend lifespan of various organisms in the laboratory. However, any improvement in aging would only provide an evolutionary benefit if the organisms were able to survive under non-ideal conditions. We have previously shown that Drosophila melanogaster carrying a loss-of-function allele of the acetyltransferase chameau (chm) has an increased healthy lifespan when fed ad libitum. Here, we show that loss of chm and reduction in its activity results in a substantial reduction in weight and a decrease in starvation resistance. This phenotype is caused by failure to properly regulate the genes and proteins required for energy storage and expenditure. The previously observed increase in survival time thus comes with the inability to prepare for and cope with nutrient stress. As the ability to survive in environments with restricted food availability is likely a stronger evolutionary driver than the ability to live a long life, chm is still present in the organism's genome despite its apparent negative effect on lifespan.
Collapse
Affiliation(s)
- Anuroop Venkateswaran Venkatasubramani
- Department of Molecular Biology, Biomedical Center Munich, Faculty of MedicineLMU MunichMartinsriedGermany
- Graduate School of Quantitative Biosciences (QBM)LMU MunichMunichGermany
| | - Toshiharu Ichinose
- Graduate School of Life SciencesTohoku UniversitySendaiJapan
- Frontier Research Institute for Interdisciplinary SciencesTohoku UniversitySendaiJapan
| | - Mai Kanno
- Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Ignasi Forne
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center MunichLMU MunichMartinsriedGermany
| | - Hiromu Tanimoto
- Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Shahaf Peleg
- Research Group Epigenetics, Metabolism and LongevityInstitute for Farm Animal BiologyDummerstorfGermany
| | - Axel Imhof
- Department of Molecular Biology, Biomedical Center Munich, Faculty of MedicineLMU MunichMartinsriedGermany
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center MunichLMU MunichMartinsriedGermany
| |
Collapse
|
35
|
Wen W, Zheng H, Li W, Huang G, Chen P, Zhu X, Cao Y, Li J, Huang X, Huang Y. Transcription factor EB: A potential integrated network regulator in metabolic-associated cardiac injury. Metabolism 2023; 147:155662. [PMID: 37517793 DOI: 10.1016/j.metabol.2023.155662] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/01/2023]
Abstract
With the worldwide pandemic of metabolic diseases, such as obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD), cardiometabolic disease (CMD) has become a significant cause of death in humans. However, the pathophysiology of metabolic-associated cardiac injury is complex and not completely clear, and it is important to explore new strategies and targets for the treatment of CMD. A series of pathophysiological disturbances caused by metabolic disorders, such as insulin resistance (IR), hyperglycemia, hyperlipidemia, mitochondrial dysfunction, oxidative stress, inflammation, endoplasmic reticulum stress (ERS), autophagy dysfunction, calcium homeostasis imbalance, and endothelial dysfunction, may be related to the incidence and development of CMD. Transcription Factor EB (TFEB), as a transcription factor, has been extensively studied for its role in regulating lysosomal biogenesis and autophagy. Recently, the regulatory role of TFEB in other biological processes, including the regulation of glucose homeostasis, lipid metabolism, etc. has been gradually revealed. In this review, we will focus on the relationship between TFEB and IR, lipid metabolism, endothelial dysfunction, oxidative stress, inflammation, ERS, calcium homeostasis, autophagy, and mitochondrial quality control (MQC) and the potential regulatory mechanisms among them, to provide a comprehensive summary for TFEB as a potential new therapeutic target for CMD.
Collapse
Affiliation(s)
- Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Haoxiao Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Weiwen Li
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Guolin Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Peng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Xiaolin Zhu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| | - Yue Cao
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Jiahuan Li
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China; The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation Research, Guangzhou, China; Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong 528308, China.
| |
Collapse
|
36
|
Chen S, Wang F, Yang G, Yuan G, Liu M, Goldman G, Harris S, Wang W, Chen Z, Mary M. Loss of Bmp2 impairs odontogenesis via dysregulating pAkt/pErk/GCN5/Dlx3/Sp7. RESEARCH SQUARE 2023:rs.3.rs-3299295. [PMID: 37790473 PMCID: PMC10543288 DOI: 10.21203/rs.3.rs-3299295/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
BMP2 signaling plays a pivotal role in odontoblast differentiation and maturation during odontogenesis. Teeth lacking Bmp2 exhibit a morphology reminiscent of dentinogenesis imperfecta (DGI), associated with mutations in dentin matrix protein 1 (DMP1) and dentin sialophosphoprotein (DSPP) genes. Mechanisms by which BMP2 signaling influences expressions of DSPP and DMP1 and contributes to DGI remain elusive. To study the roles of BMP2 in dentin development, we generated Bmp2 conditional knockout (cKO) mice. Through a comprehensive approach involving RNA-seq, immunohistochemistry, promoter activity, ChIP, and Re-ChIP, we investigated downstream targets of Bmp2. Notably, the absence of Bmp2 in cKO mice led to dentin insufficiency akin to DGI. Disrupted Bmp2 signaling was linked to decreased expression of Dspp and Dmp1, as well as alterations in intracellular translocation of transcription factors Dlx3 and Sp7. Intriguingly, upregulation of Dlx3, Dmp1, Dspp, and Sp7, driven by BMP2, fostered differentiation of dental mesenchymal cells and biomineralization. Mechanistically, BMP2 induced phosphorylation of Dlx3, Sp7, and histone acetyltransferase GCN5 at Thr and Tyr residues, mediated by Akt and Erk42/44 kinases. This phosphorylation facilitated protein nuclear translocation, promoting interactions between Sp7 and Dlx3, as well as with GCN5 on Dspp and Dmp1 promoters. The synergy between Dlx3 and Sp7 bolstered transcription of Dspp and Dmp1. Notably, BMP2-driven GCN5 acetylated Sp7 and histone H3, while also recruiting RNA polymerase II to Dmp1 and Dspp chromatins, enhancing their transcriptions. Intriguingly, BMP2 suppressed the expression of histone deacetylases. we unveil hitherto uncharted involvement of BMP2 in dental cell differentiation and dentine development through pAkt/pErk42/44/Dlx3/Sp7/GCN5/Dspp/Dmp1.
Collapse
Affiliation(s)
- Shuo Chen
- UT Health Science Center at San Antonio
| | | | | | | | - Mengmeng Liu
- School of Dentistry, the University of Texas Health Science Center at San Antonio
| | - Graham Goldman
- School of Dentistry, the University of Texas Health Science Center at San Antonio
| | | | | | - Zhi Chen
- Wuhan University School and Hospital of Stomatology
| | | |
Collapse
|
37
|
Li J, Cao Y, Yang Y, Ma H, Zhao J, Zhang Y, Liu N. Quantitative Acetylomics Reveals Substrates of Lysine Acetyltransferase GCN5 in Adult and Aging Drosophila. J Proteome Res 2023; 22:2909-2924. [PMID: 37545086 DOI: 10.1021/acs.jproteome.3c00247] [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: 08/08/2023]
Abstract
Protein lysine acetylation is a dynamic post-translational modification (PTM) that regulates a wide spectrum of cellular events including aging. General control nonderepressible 5 (GCN5) is a highly conserved lysine acetyltransferase (KAT). However, the acetylation substrates of GCN5 in vivo remain poorly studied, and moreover, how lysine acetylation changes with age and the contribution of KATs to aging remain to be addressed. Here, using Drosophila, we perform label-free quantitative acetylomic analysis, identifying new substrates of GCN5 in the adult and aging process. We further characterize the dynamics of protein acetylation with age, which exhibits a trend of increase. Since the expression of endogenous fly Gcn5 progressively increases during aging, we reason that, by combining the substrate analysis, the increase in acetylation with age is triggered, at least in part, by GCN5. Collectively, our study substantially expands the atlas of GCN5 substrates in vivo, provides a resource of protein acetylation that naturally occurs with age, and demonstrates how individual KAT contributes to the aging acetylome.
Collapse
Affiliation(s)
- Jingshu Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ye Cao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yun Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huanhuan Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Zhao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- Shanghai Key Laboratory of Aging Studies, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
| | - Nan Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
- Shanghai Key Laboratory of Aging Studies, 100 Hai Ke Rd., Pudong, Shanghai 201210, China
| |
Collapse
|
38
|
Zhang L, Li Z, Zhang L, Qin Y, Yu D. Dissecting the multifaced function of transcription factor EB (TFEB) in human diseases: From molecular mechanism to pharmacological modulation. Biochem Pharmacol 2023; 215:115698. [PMID: 37482200 DOI: 10.1016/j.bcp.2023.115698] [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: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The transcription factor EB (TFEB) is a transcription factor of the MiT/TFE family that translocations from the cytoplasm to the nucleus in response to various stimuli, including lysosomal stress and nutrient starvation. By activating genes involved in lysosomal function, autophagy, and lipid metabolism, TFEB plays a crucial role in maintaining cellular homeostasis. Dysregulation of TFEB has been implicated in various diseases, including cancer, neurodegenerative diseases, metabolic diseases, cardiovascular diseases, infectious diseases, and inflammatory diseases. Therefore, modulating TFEB activity with agonists or inhibitors may have therapeutic potential. In this review, we reviewed the recently discovered regulatory mechanisms of TFEB and their impact on human diseases. Additionally, we also summarize the existing TFEB inhibitors and agonists (targeted and non-targeted) and discuss unresolved issues and future research directions in the field. In summary, this review sheds light on the crucial role of TFEB, which may pave the way for its translation from basic research to practical applications, bringing us closer to realizing the full potential of TFEB in various fields.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, 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.
| | - Yuan Qin
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China.
| | - Dongke Yu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
39
|
Romeo MA, Gilardini Montani MS, Santarelli R, Benedetti R, Arena A, Cirone M. Acetylation increases expression, interaction with TRAPPC4 and surface localization of PD-L1. Discov Oncol 2023; 14:152. [PMID: 37603071 PMCID: PMC10442048 DOI: 10.1007/s12672-023-00766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/09/2023] [Indexed: 08/22/2023] Open
Abstract
PD-L1 is an immune checkpoint inhibitor, whose surface expression may be exploited by cancer cells to escape T cell-mediated immune recognition. PD-L1 expression and nuclear localization can be affected by epigenetic modifications, such as acetylation. In this study, we showed that VPA, a class I/IIa HDAC inhibitor, upregulated PD-L1 expression on the surface of pancreatic cancer cells. To this effect contributed the increased transcription, in correlation with histone acetylation of the PD-L1 gene and the acetylation of PD-L1 protein, which led to an increased interaction with TRAPPC4, molecule involved in PD-L1 recycling to the cell membrane. Interestingly, the BRD4 inhibitor JQ-1, counteracted PD-L1 transcription and reduced its surface expression, suggesting that such a combination could improve the outcome of VPA treatment, also because it increased the cytotoxic effect of VPA. Also considering that this HDACi did not upregulate PD-L2 and that the supernatant of VPA-treated cancer cells did not increase PD-L1 expression on the surface of macrophages exposed to it.
Collapse
Affiliation(s)
- Maria Anele Romeo
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | | | - Roberta Santarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Rossella Benedetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Andrea Arena
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
40
|
Zhang Z, Zhu Q. WD Repeat and HMG Box DNA Binding Protein 1: An Oncoprotein at the Hub of Tumorigenesis and a Novel Therapeutic Target. Int J Mol Sci 2023; 24:12494. [PMID: 37569867 PMCID: PMC10420296 DOI: 10.3390/ijms241512494] [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: 07/21/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
WD repeat and HMG-box DNA binding protein 1 (WDHD1) is a highly conserved gene from yeast to humans. It actively participates in DNA replication, playing a crucial role in DNA damage repair and the cell cycle, contributing to centromere formation and sister chromosome segregation. Notably, several studies have implicated WDHD1 in the development and progression of diverse tumor types, including esophageal carcinoma, pulmonary carcinoma, and breast carcinoma. Additionally, the inhibitor of WDHD1 has been found to enhance radiation sensitivity, improve drug resistance, and significantly decrease tumor cell proliferation. This comprehensive review aims to provide an overview of the molecular structure, biological functions, and regulatory mechanisms of WDHD1 in tumors, thereby establishing a foundation for future investigations and potential clinical applications of WDHD1.
Collapse
Affiliation(s)
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China;
| |
Collapse
|
41
|
Hu JH, Li SY, Yu LH, Guan ZR, Jiang YP, Hu D, Wang HJ, Zhao LP, Zhou ZH, Yan YX, Xie T, Huang ZH, Lou JS. TFEB: a double-edged sword for tumor metastasis. J Mol Med (Berl) 2023; 101:917-929. [PMID: 37328669 DOI: 10.1007/s00109-023-02337-0] [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: 02/18/2023] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023]
Abstract
Transcription factor EB, a member of the microphthalmia-associated transcription factor (MiTF/TFE) family, is a master regulator of autophagy, lysosome biogenesis, and TAMs. Metastasis is one of the main reasons for the failure of tumor therapy. Studies on the relationship between TFEB and tumor metastasis are contradictory. On the positive side, TFEB mainly affects tumor cell metastasis via five aspects, including autophagy, epithelial-mesenchymal transition (EMT), lysosomal biogenesis, lipid metabolism, and oncogenic signaling pathways; on the negative side, TFEB mainly affects tumor cell metastasis in two aspects, including tumor-associated macrophages (TAMs) and EMT. In this review, we described the detailed mechanism of TFEB-mediated regulation of metastasis. In addition, we also described the activation and inactivation of TFEB in several aspects, including the mTORC1 and Rag GTPase systems, ERK2, and AKT. However, the exact process by which TFEB regulates tumor metastasis remains unclear in some pathways, which requires further studies.
Collapse
Affiliation(s)
- Jun-Hu Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Shou-Ye Li
- College of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, 311300, China
- Zhejiang Eyoung Pharmaceutical Research and Development Center, Hangzhou, Zhejiang, 311258, China
| | - Li-Hua Yu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zhen-Rong Guan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Ya-Ping Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Die Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Hao-Jie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Li-Ping Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zhao-Huang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Ya-Xin Yan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Zhi-Hui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Jian-Shu Lou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
42
|
Wang P, Zhao C, Zhou H, Huang X, Ying H, Zhang S, Pan Y, Zhu H. Dysregulation of Histone Deacetylases Inhibits Trophoblast Growth during Early Placental Development Partially through TFEB-Dependent Autophagy-Lysosomal Pathway. Int J Mol Sci 2023; 24:11899. [PMID: 37569278 PMCID: PMC10418899 DOI: 10.3390/ijms241511899] [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: 06/23/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Dysregulated biological behaviors of trophoblast cells can result in recurrent spontaneous abortion (RSA)-whose underlying etiology still remains insufficient. Autophagy, a conserved intracellular physiological process, is precisely monitored throughout whole pregnancy. Although the exact mechanism or role remains elusive, epigenetic modification has emerged as an important process. Herein, we found that a proportion of RSA patients exhibited higher levels of autophagy in villus tissues compared to controls, accompanied with impaired histone deacetylase (HDAC) expression. The purpose of this study is to explore the connection between HDACs and autophagy in the pathological course of RSA. Mechanistically, using human trophoblast cell models, treatment with HDAC inhibitor (HDACI)-trichostatin A (TSA) can induce autophagy by promoting nuclear translocation and transcriptional activity of the central autophagic regulator transcription factor EB (TFEB). Specifically, overactivated autophagy is involved in the TSA-driven growth inhibition of trophoblast, which can be partially reversed by the autophagy inhibitor chloroquine (CQ) or RNA interference of TFEB. In summary, our results reveal that abnormal acetylation and autophagy levels during early gestation may be associated with RSA and suggest the potential novel molecular target TFEB for RSA treatment.
Collapse
Affiliation(s)
- Peixin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Chenqiong Zhao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Hanjing Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Xiaona Huang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Hanqi Ying
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Yibin Pan
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| | - Haiyan Zhu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou 310016, China
| |
Collapse
|
43
|
Zhang C, Chen H, Rodriguez Y, Ma X, Swerdlow RH, Zhang J, Ding WX. A perspective on autophagy and transcription factor EB in Alcohol-Associated Alzheimer's disease. Biochem Pharmacol 2023; 213:115576. [PMID: 37127251 PMCID: PMC11009931 DOI: 10.1016/j.bcp.2023.115576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
Alzheimer's disease (AD) is the most common form of progressive dementia and there is no truly efficacious treatment. Accumulating evidence indicates that impaired autophagic function for removal of damaged mitochondria and protein aggregates such as amyloid and tau protein aggregates may contribute to the pathogenesis of AD. Epidemiologic studies have implicated alcohol abuse in promoting AD, yet the underlying mechanisms are poorly understood. In this review, we discuss mechanisms of selective autophagy for mitochondria and protein aggregates and how these mechanisms are impaired by aging and alcohol consumption. We also discuss potential genetic and pharmacological approaches for targeting autophagy/mitophagy, as well as lysosomal and mitochondrial biogenesis, for the potential prevention and treatment of AD.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hao Chen
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Russell H Swerdlow
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jianhua Zhang
- Department of Pathology, Division of Molecular Cellular Pathology, University of Alabama at Birmingham, 901 19th street South, Birmingham, AL 35294, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Internal Medicine, Division of Gastroenterology, Hepatology & Motility, The University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
44
|
Zhou Y, Guan J, Meng G, Fan W, Ge C, Niu C, Cheng Y, Fu Y, Lu Y, Wei Y. The RagA GTPase protects young egg chambers in Drosophila. Cell Rep 2023; 42:112631. [PMID: 37302067 DOI: 10.1016/j.celrep.2023.112631] [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: 12/15/2022] [Revised: 04/18/2023] [Accepted: 05/24/2023] [Indexed: 06/13/2023] Open
Abstract
The preservation of female fertility under unfavorable conditions is essential for animal reproduction. Inhibition of the target of rapamycin complex 1 (TORC1) is indispensable for Drosophila young egg chamber maintenance under nutrient starvation. Here, we show that knockdown of RagA results in young egg chamber death independent of TORC1 hyperactivity. RagA RNAi ovaries have autolysosomal acidification and degradation defects, which make the young egg chambers sensitive to autophagosome augmentation. Meanwhile, RagA RNAi ovaries have nuclear-localized Mitf, which promotes autophagic degradation and protects young egg chambers under stress. Interestingly, GDP-bound RagA rescues autolysosome defects, while GTP-bound RagA rescues Mitf nuclear localization in RagA RNAi young egg chambers. Moreover, Rag GTPase activity, rather than TORC1 activity, controls Mitf cellular localization in the Drosophila germ line. Our work suggests that RagA separately controls autolysosomal acidification and Mitf activity in the Drosophila young egg chambers.
Collapse
Affiliation(s)
- Ying Zhou
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Jianwen Guan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Guoqiang Meng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Weikang Fan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Churui Ge
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Chunmei Niu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Yang Cheng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Yuanyuan Fu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Yingying Lu
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Youheng Wei
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
45
|
Du J, Liu L, Fan H, Yu Y, Luo Y, Gu F, Yu H, Liao X. Anthocyanins improve liver fibrosis in mice by regulating the autophagic flux level of hepatic stellate cells by mmu_circ_0000623. Food Sci Nutr 2023; 11:3002-3018. [PMID: 37324880 PMCID: PMC10261807 DOI: 10.1002/fsn3.3281] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 06/17/2023] Open
Abstract
Liver fibrosis is a key step in the progression of various chronic liver diseases to liver cirrhosis and even liver cancer, it is also an important link affecting prognosis. Therefore, this study aimed to investigate the therapeutic effect of anthocyanins on liver fibrosis and the molecular mechanism of mmu_circ_0000623 in anthocyanin therapy. In this study, CCL4 was used to build a mouse liver fibrosis model, and the treatment groups were treated with 100 and 200 mg/kg of anthocyanins daily by gavage. Liver fibrosis indicators, macrophage polarization markers, and liver injury markers were further detected by real-time quantitative PCR (qRT-PCR), western blotting (WB), and enzyme-linked immunosorbent assay. Morphological verification of liver injury in different treatment groups was performed by histopathological method. A mouse hepatic stellate cell (HSC) model and a mouse liver fibrosis model were constructed to verify the expression of circ_0000623, miR-351-5p, and TFEB. Transfected with mRFP-GFP-LC3 to detect the autophagic flux of HSCs. We found that 100 mg/kg or 200 mg/kg of anthocyanins could significantly reduce the degree of liver fibrosis in mice. In addition, anthocyanins can inhibit the proliferation, activation, and migration ability of HSCs. circ_0000623 was lowly expressed in mice with liver fibrosis, and anthocyanin treatment could promote its increased expression. Further testing found that anthocyanins could reverse the blocked autophagic flux induced by PDGF or CCL4. This effect is achieved by regulating the expression of TFEB by competitive adsorption of miR-351-5p. Anthocyanins could treat liver fibrosis by modulating circ_0000623/miR-351-5p/TFEB-mediated changes in HSC autophagic flux.
Collapse
Affiliation(s)
- Jinhui Du
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | - Likun Liu
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | - Haiqing Fan
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | - Yue Yu
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | - Yilin Luo
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | - Fang Gu
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | - Hui Yu
- Department of Medical ImagingThe Affiliated Hospital of Guizhou Medical UniversityGuiyang CityChina
| | | |
Collapse
|
46
|
Wang Z, Yang C, Guan D, Li J, Zhang H. Cellular proteins act as surfactants to control the interfacial behavior and function of biological condensates. Dev Cell 2023:S1534-5807(23)00157-0. [PMID: 37098348 DOI: 10.1016/j.devcel.2023.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/02/2023] [Accepted: 04/01/2023] [Indexed: 04/27/2023]
Abstract
Interfacial tension governs the behaviors and physiological functions of multiple biological condensates during diverse biological processes. Little is known about whether there are cellular surfactant factors that regulate the interfacial tension and functions of biological condensates within physiological environments. TFEB, a master transcription factor that controls expression of autophagic-lysosomal genes, assembles into transcriptional condensates to control the autophagy-lysosome pathway (ALP). Here, we show that interfacial tension modulates the transcriptional activity of TFEB condensates. MLX, MYC, and IPMK act as synergistic surfactants to decrease the interfacial tension and consequent DNA affinity of TFEB condensates. The interfacial tension of TFEB condensates is quantitatively correlated to their DNA affinity and subsequent ALP activity. The interfacial tension and DNA affinity of condensates formed by TAZ-TEAD4 are also regulated by the synergistic surfactant proteins RUNX3 and HOXA4. Our results indicate that the interfacial tension and functions of biological condensates can be controlled by cellular surfactant proteins in human cells.
Collapse
Affiliation(s)
- Zheng Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China.
| | - Chun Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Dongshi Guan
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Jiaqi Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
47
|
Shillingford JM, Shayman JA. Functional TFEB activation characterizes multiple models of renal cystic disease and loss of polycystin-1. Am J Physiol Renal Physiol 2023; 324:F404-F422. [PMID: 36794754 PMCID: PMC10069964 DOI: 10.1152/ajprenal.00237.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Polycystic kidney disease is a disorder of renal epithelial growth and differentiation. Transcription factor EB (TFEB), a master regulator of lysosome biogenesis and function, was studied for a potential role in this disorder. Nuclear translocation and functional responses to TFEB activation were studied in three murine models of renal cystic disease, including knockouts of folliculin, folliculin interacting proteins 1 and 2, and polycystin-1 (Pkd1) as well as in mouse embryonic fibroblasts lacking Pkd1 and three-dimensional cultures of Madin-Darby canine kidney cells. Nuclear translocation of Tfeb characterized cystic but not noncystic renal tubular epithelia in all three murine models as both an early and sustained response to cyst formation. Epithelia expressed elevated levels of Tfeb-dependent gene products, including cathepsin B and glycoprotein nonmetastatic melanoma protein B. Nuclear Tfeb translocation was observed in mouse embryonic fibroblasts lacking Pkd1 but not wild-type fibroblasts. Pkd1 knockout fibroblasts were characterized by increased Tfeb-dependent transcripts, lysosomal biogenesis and repositioning, and increased autophagy. The growth of Madin-Darby canine kidney cell cysts was markedly increased following exposure to the TFEB agonist compound C1, and nuclear Tfeb translocation was observed in response to both forskolin and compound C1 treatment. Nuclear TFEB also characterized cystic epithelia but not noncystic tubular epithelia in human patients with autosomal dominant polycystic kidney disease. Noncanonical activation of TFEB is characteristic of cystic epithelia in multiple models of renal cystic disease including those associated with loss of Pkd1. Nuclear TFEB translocation is functionally active in these models and may be a component of a general pathway contributing to cystogenesis and growth.NEW & NOTEWORTHY Changes in epithelial cell metabolism are important in renal cyst development. The role of TFEB, a transcriptional regulator of lysosomal function, was explored in several models of renal cystic disease and human ADPKD tissue sections. Nuclear TFEB translocation was uniformly observed in cystic epithelia in each model of renal cystic disease examined. TFEB translocation was functionally active and associated with lysosomal biogenesis and perinuclear repositioning, increased TFEB-associated protein expression, and activation of autophagic flux. Compound C1, a TFEB agonist, promoted cyst growth in 3-D cultures of MDCK cells. Nuclear TFEB translocation is an underappreciated signaling pathway for cystogenesis that may represent a new paradigm for cystic kidney disease.
Collapse
Affiliation(s)
- Jonathan M Shillingford
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan, United States
| | - James A Shayman
- Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
48
|
Martina JA, Jeong E, Puertollano R. p38 MAPK-dependent phosphorylation of TFEB promotes monocyte-to-macrophage differentiation. EMBO Rep 2023; 24:e55472. [PMID: 36507874 PMCID: PMC9900348 DOI: 10.15252/embr.202255472] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
The transcription factor EB (TFEB) regulates energy homeostasis and cellular response to a wide variety of stress conditions, including nutrient deprivation, oxidative stress, organelle damage, and pathogens. Here we identify S401 as a novel phosphorylation site within the TFEB proline-rich domain. Phosphorylation of S401 increases significantly in response to oxidative stress, UVC light, growth factors, and LPS, whereas this increase is prevented by p38 MAPK inhibition or depletion, revealing a new role for p38 MAPK in TFEB regulation. Mutation of S401 in THP1 cells demonstrates that the p38 MAPK/TFEB pathway plays a particularly relevant role during monocyte differentiation into macrophages. TFEB-S401A monocytes fail to upregulate the expression of multiple immune genes in response to PMA-induced differentiation, including critical cytokines, chemokines, and growth factors. Polarization of M0 macrophages into M1 inflammatory macrophages is also aberrant in TFEB-S401A cells. These results indicate that TFEB-S401 phosphorylation links differentiation signals to the transcriptional control of monocyte differentiation.
Collapse
Affiliation(s)
- José A Martina
- Cell and Developmental Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Eutteum Jeong
- Cell and Developmental Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Rosa Puertollano
- Cell and Developmental Biology CenterNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
49
|
Abstract
Post-translational modifications, such as phosphorylation, ubiquitination and acetylation, play crucial roles in the regulation of autophagy. Acetylation has emerged as an important regulatory mechanism for autophagy. Acetylation regulates autophagy initiation and autophagosome formation by targeting core components of the ULK1 complex, the BECN1-PIK3C3 complex, and the LC3 lipidation system. Recent studies have shown that acetylation occurs on the key proteins participating in autophagic cargo assembly and autophagosome-lysosome fusion, such as SQSTM1/p62 and STX17. In addition, acetylation controls autophagy at the transcriptional level by targeting histones and the transcription factor TFEB. Here, we review the current knowledge on acetylation of autophagy proteins and their regulations and functions in the autophagy pathway with focus on recent findings.Abbreviations : ACAT1: acetyl-CoA acetyltransferase 1; ACSS2: acyl-CoA synthetase short chain family member 2; AMPK: AMP-activated protein kinase; ATG: autophagy-related; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CCAR2/DBC1: cell cycle and apoptosis regulator 2; BECN1: beclin 1; CMA: chaperone-mediated autophagy; CREBBP/CBP: CREB binding protein; EP300/p300: E1A binding protein p300; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GSK3: glycogen synthase kinase 3; HDAC6: histone deacetylase 6; HSPA8/HSC70: heat shock protein family A (Hsp70) member 8; KAT2A/GCN5: lysine acetyltransferase 2A; KAT2B/PCAF: lysine acetyltransferase 2B; KAT5/TIP60: lysine acetyltransferase 5; KAT8/MOF: lysine acetyltransferase 8; LAMP2A: lysosomal associated membrane protein 2A; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PD: Parkinson disease; PE: phosphatidylethanolamine; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PKM2: pyruvate kinase M1/2; PtdIns3P: phosphatidylinositol-3-phosphate; PTM: post-translational modification; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RUBCN/Rubicon: rubicon autophagy regulator; RUBCNL/Pacer: rubicon like autophagy enhancer; SIRT1: sirtuin 1; SNAP29: synaptosome associated protein 29; SNARE: soluble N-ethylamide-sensitive factor attachment protein receptor; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TFEB: transcription factor EB; TP53/p53: tumor protein p53; TP53INP2/DOR: tumor protein p53 inducible nuclear protein 2; UBA: ubiquitin-associated; ULK1: unc-51 like autophagy activating kinase 1; VAMP8: vesicle associated membrane protein 8; WIPI2: WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Yinfeng Xu
- Laboratory of Basic Biology, Hunan First Normal University, Changsha, Hunan, China
| | - Wei Wan
- Department of Biochemistry, and Department of Thoracic Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,CONTACT Wei Wan Research Building B, Room 716, Zhejiang University School of Medicine, 866 Yu-Hang-Tang Road, Hangzhou, Zhejiang310058, China
| |
Collapse
|
50
|
Shu F, Xiao H, Li QN, Ren XS, Liu ZG, Hu BW, Wang HS, Wang H, Jiang GM. Epigenetic and post-translational modifications in autophagy: biological functions and therapeutic targets. Signal Transduct Target Ther 2023; 8:32. [PMID: 36646695 PMCID: PMC9842768 DOI: 10.1038/s41392-022-01300-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/19/2022] [Accepted: 12/18/2022] [Indexed: 01/17/2023] Open
Abstract
Autophagy is a conserved lysosomal degradation pathway where cellular components are dynamically degraded and re-processed to maintain physical homeostasis. However, the physiological effect of autophagy appears to be multifaced. On the one hand, autophagy functions as a cytoprotective mechanism, protecting against multiple diseases, especially tumor, cardiovascular disorders, and neurodegenerative and infectious disease. Conversely, autophagy may also play a detrimental role via pro-survival effects on cancer cells or cell-killing effects on normal body cells. During disorder onset and progression, the expression levels of autophagy-related regulators and proteins encoded by autophagy-related genes (ATGs) are abnormally regulated, giving rise to imbalanced autophagy flux. However, the detailed mechanisms and molecular events of this process are quite complex. Epigenetic, including DNA methylation, histone modifications and miRNAs, and post-translational modifications, including ubiquitination, phosphorylation and acetylation, precisely manipulate gene expression and protein function, and are strongly correlated with the occurrence and development of multiple diseases. There is substantial evidence that autophagy-relevant regulators and machineries are subjected to epigenetic and post-translational modulation, resulting in alterations in autophagy levels, which subsequently induces disease or affects the therapeutic effectiveness to agents. In this review, we focus on the regulatory mechanisms mediated by epigenetic and post-translational modifications in disease-related autophagy to unveil potential therapeutic targets. In addition, the effect of autophagy on the therapeutic effectiveness of epigenetic drugs or drugs targeting post-translational modification have also been discussed, providing insights into the combination with autophagy activators or inhibitors in the treatment of clinical diseases.
Collapse
Affiliation(s)
- Feng Shu
- grid.452859.70000 0004 6006 3273Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong China
| | - Han Xiao
- grid.452859.70000 0004 6006 3273Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong China
| | - Qiu-Nuo Li
- grid.452859.70000 0004 6006 3273Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong China
| | - Xiao-Shuai Ren
- grid.452859.70000 0004 6006 3273Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong China
| | - Zhi-Gang Liu
- grid.284723.80000 0000 8877 7471Cancer Center, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong China
| | - Bo-Wen Hu
- grid.452859.70000 0004 6006 3273Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong China
| | - Hong-Sheng Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Hao Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Guan-Min Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|