1
|
Ji HL, Liu C, Zhang JJ, Lin L, Yang Q, Yang Y, Dong CC, He YB, Shao CW. Molecular cloning, expression, and functional analyses of plasmanylethanolamine desaturase gene of Takifugu rubripes. Gene 2025; 953:149242. [PMID: 39800193 DOI: 10.1016/j.gene.2025.149242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
The aging population has led to a significant increase in neurodegenerative diseases, particularly Alzheimer's disease (AD), which adversely affects the quality of life and longevity of the elderly. Abnormal plasmalogen metabolism plays a crucial role in the pathogenesis of AD. This study focused on tmem189, a key gene involved in plasmalogen synthesis. We successfully cloned and characterized the open reading frame (ORF) of tmem189 in Takifugu rubripes, revealing that it encodes a protein consisting of 275 amino acids. Notably, tmem189 expression was found to be higher in brain compared to other tissues. We transfected a GFP-tagged eukaryotic expression vector into 293 T cells, confirming successful expression of tmem189 with increased expression levels. Additionally, liquid chromatography-mass spectrometry (LC-MS) analysis demonstrated that tmem189 promotes plasmalogen synthesis in the transfected 293T cells. Our findings suggest that tmem189 could serve as a potential target for the treatment of neurodegenerative diseases, providing new insights into the promotion of plasmalogen synthesis.
Collapse
Affiliation(s)
- Hong-Long Ji
- College of Fisheries, Zhejiang Ocean University, Zhoushan 316022, China; State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China.
| | - Chang Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Jing-Jing Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Lei Lin
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Qi Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Yu Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Cai-Chao Dong
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Yang-Bin He
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| | - Chang-Wei Shao
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, China.
| |
Collapse
|
2
|
Xu J, Weng C, Zhang Y, Zhao Q, Chen J, Pan S, Wang Y, Zhang R, Wang Y, Zhu W, Cao M, Zu D, Zhang S, Xu Z, Hu C, Cheng X. GPX4 knockdown suppresses M2 macrophage polarization in gastric cancer by modulating kynurenine metabolism. Theranostics 2025; 15:5826-5845. [PMID: 40365295 PMCID: PMC12068284 DOI: 10.7150/thno.108817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Glutathione peroxidase 4 (GPX4), an important factor regulating redox homeostasis, plays an important role in tumor microenvironment and progression. However, the role of GPX4 in gastric cancer (GC) is unclear. Methods: Spectral flow cytometry and multiplex immunohistochemistry were employed to assess the correlation between GPX4 expression and immune cell infiltration. Metabolomics analysis of conditioned media from GPX4 knockdown NUGC3 cells identified metabolic alterations. Additionally, both in vitro and in vivo functional studies were conducted to elucidate the mechanistic role of GPX4 in regulating the tumor microenvironment and progression. Results: Knockdown of GPX4 in GC cells inhibited tumor growth, enhanced CD8+ T cell infiltration, and suppressed the polarization of tumor-associated macrophages (TAMs) toward the pro-tumor M2 phenotype. Multiplex immunohistochemistry revealed a positive correlation between GPX4 expression and M2 macrophage infiltration in clinical samples from patients with GC. Metabolomics revealed that GPX4 knockdown regulate kynurenine metabolism pathway. Furthermore, mechanistic studies reveal that GPX4 silencing elevates lipid peroxidation, triggering the conversion of KYNU ubiquitin chain modifications from K48 to K63. Such ubiquitination remodeling stabilizes KYNU expression (a key kynurenine-metabolizing enzyme), reduces kynurenine accumulation, and ultimately reprograms TAM polarization to enhance antitumor immunity. We also identified that the K96 and K163 sites are important for KYNU's modification by K48 and K63 ubiquitin chains. Conclusion: Our study not only affirm the role of GPx4 in GC progression but also highlight it as a promising target for reshaping the immune microenvironment.
Collapse
Affiliation(s)
- Jingli Xu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chunyan Weng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Qianyu Zhao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jiahui Chen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Siwei Pan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yan Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ruolan Zhang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yuqi Wang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Weiwei Zhu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Mengxuan Cao
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Dan Zu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Shengjie Zhang
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
3
|
Huang L, Xu K, Yang Q, Ding Z, Shao Z, Li E. ANXA2 in cancer: aberrant regulation of tumour cell apoptosis and its immune interactions. Cell Death Discov 2025; 11:174. [PMID: 40234383 PMCID: PMC12000292 DOI: 10.1038/s41420-025-02469-x] [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: 12/08/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Annexin A2 (ANXA2) is a multifunctional protein that binds to calcium and phospholipids and plays a critical role in various pathological conditions, including cancer and inflammation. Recently, there has been increasing recognition of the significant role of ANXA2 in inhibiting apoptosis and promoting immune evasion in tumour cells. Therefore, a deep understanding of the regulatory mechanisms of ANXA2 in tumour cell apoptosis and its relationship with immune evasion can provide new targets for cancer therapy. This review summarizes the role and mechanisms of ANXA2 in regulating apoptosis in tumour cells, the connection between apoptosis regulation and tumour immunity, and the potential role of ANXA2 in therapy resistance.
Collapse
Affiliation(s)
- Le Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Kailing Xu
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Qingping Yang
- Department of Reproductive Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai zheng Street, Nanchang, Jiangxi, 330006, China
| | - Zijun Ding
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhenduo Shao
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Enliang Li
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, Jiangxi, China.
| |
Collapse
|
4
|
Racioppo B, Pechalrieu D, Abegg D, Dwyer B, Ramseier NT, Hu YS, Adibekian A. Chemoproteomics-Enabled De Novo Proteolysis Targeting Chimera Discovery Platform Identifies a Metallothionein Degrader to Probe Its Role in Cancer. J Am Chem Soc 2025; 147:7817-7828. [PMID: 39989026 DOI: 10.1021/jacs.4c17827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Proteolysis targeting chimeras (PROTACs) represent powerful tools to modulate the activity of classically "undruggable" proteins, but their application has been limited to known ligands and a few select protein classes. Herein, we present our chemoproteomic strategy for simultaneous de novo discovery of novel degraders and ligands for challenging and previously "undruggable" targets. Using comparative PROTAC versus ligand global proteomics analyses, we rapidly identify proteins selectively downregulated by several "untargeted" PROTACs containing a VHL E3 ligase recruiter and various covalent and noncovalent ligands. We showcase our approach by identifying a first-in-class PROTAC for metallothionein 2A (MT2A), a small, cysteine-rich, metal-binding protein implicated in heavy metal detoxification, zinc homeostasis, and cellular invasion. Notably, isoform-specific MT overexpression has been shown to augment cellular migration and invasion across several cancer cell lines, although the precise mechanisms are unknown due to insufficient tools to study MTs. We show that optimized PROTAC AA-BR-157 covalently binds conserved C44, degrades overexpressed MT2A with nanomolar potency, and reduces the migration and invasion of MDA-MB-231 cells. We further demonstrate a time-dependent increase in intracellular zinc levels following MT2A degradation as well as downregulation of protein diaphanous homolog 3 (DIAPH3), a positive regulator of actin and cell motility. Super-resolution imaging of MDA-MB-231 cells shows that the downregulation of MT2A and DIAPH3 inhibits cell polarization and thereby migration, suggesting that MT2A may regulate motility via DIAPH3-dependent cytoskeletal remodeling. In summary, our strategy enables the de novo discovery of PROTACs and ligands for novel disease-related targets and lays the groundwork for expansion of the druggable proteome.
Collapse
Affiliation(s)
- Brittney Racioppo
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor Street, Chicago, Illinois 60607, United States
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Road, La Jolla, California 92037, United States
| | - Dany Pechalrieu
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor Street, Chicago, Illinois 60607, United States
| | - Daniel Abegg
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor Street, Chicago, Illinois 60607, United States
| | - Brendan Dwyer
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Road, La Jolla, California 92037, United States
| | - Neal Thomas Ramseier
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor Street, Chicago, Illinois 60607, United States
| | - Ying S Hu
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor Street, Chicago, Illinois 60607, United States
| | - Alexander Adibekian
- Department of Chemistry, University of Illinois Chicago, 845 W Taylor Street, Chicago, Illinois 60607, United States
- University of Illinois Cancer Center, 818 South Wolcott Avenue, Chicago, Illinois 60612, United States
- UICentre, University of Illinois Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Department of Pharmaceutical Sciences, University of Illinois Chicago, 833 S Wood Street, Chicago, Illinois 60607, United States
- Department of Biochemistry and Molecular Genetics, University of Illinois, 900 S Ashland Ave, Chicago, Illinois 60607, United States
| |
Collapse
|
5
|
Qiu G, Song C, Lou M, Lin J. Decoding the Expressions, Immune Relevance, and Prognostic Values of Ferroptosis Gene TMEM189: A Pan-cancer Analysis. Curr Cancer Drug Targets 2025; 25:520-537. [PMID: 38934284 DOI: 10.2174/0115680096308701240605114342] [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: 02/05/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND TMEM189 is a recently discovered transmembrane protein involved in ether glycerophospholipid synthesis and ferroptosis regulation. However, its role in tumors is not well understood. OBJECTIVE This study aimed to elucidate the oncogenic effects and prognostic values of TMEM189 in tumors. METHODS We performed a pan-cancer analysis of TMEM189 using various databases, bioinformatics and statistical tools, and tissue microarray analysis. RESULTS TMEM189 was upregulated in tumors compared to normal tissues. High TMEM189 expression was found to be linked to reduced promoter methylation. Moreover, TMEM189 exhibited a negative correlation with immunogenic markers, immune cell infiltration, and expression of Immune Checkpoint Genes (ICGs) in most cancers, implicating its immunosuppressive role in tumor microenvironments (TME). The genes that interact and are similar to TMEM189 were involved in hotspot signaling pathways in pan-cancer. TMEM189 overexpression was found to be usually associated with poor prognosis, especially an independent prognostic risk factor for BLCA, BRCA, LUAD, MESO, LIHC, and SKCM. CONCLUSION TMEM189 is overexpressed and exerts immunosuppressive effects in many tumors with a significant association with poor prognosis, suggesting its potential as a therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Guanzhong Qiu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Chaoli Song
- Department of Neurosurgery, Western Theater Command Air Force Hospital, Chengdu, 600021, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Jing Lin
- Department of Neurosurgery, Western Theater Command Air Force Hospital, Chengdu, 600021, China
- Department of Health Statistics, Naval Medical University, Shanghai, 200003, China
| |
Collapse
|
6
|
Arroyo AB, Tyrkalska SD, Bastida-Martínez E, Monera-Girona AJ, Cantón-Sandoval J, Bernal-Carrión M, García-Moreno D, Elías-Arnanz M, Mulero V. Peds1 deficiency in zebrafish results in myeloid cell apoptosis and exacerbated inflammation. Cell Death Discov 2024; 10:388. [PMID: 39209813 PMCID: PMC11362147 DOI: 10.1038/s41420-024-02141-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Plasmalogens are glycerophospholipids with a vinyl ether bond that confers unique properties. Recent identification of the gene encoding PEDS1, the desaturase generating the vinyl ether bond, enables evaluation of the role of plasmalogens in health and disease. Here, we report that Peds1-deficient zebrafish larvae display delayed development, increased basal inflammation, normal hematopoietic stem and progenitor cell emergence, and cell-autonomous myeloid cell apoptosis. In a sterile acute inflammation model, Peds1-deficient larvae exhibited impaired inflammation resolution and tissue regeneration, increased interleukin-1β and NF-κB expression, and elevated ROS levels at the wound site. Abnormal immune cell recruitment, neutrophil persistence, and fewer but predominantly pro-inflammatory macrophages were observed. Chronic skin inflammation worsened in Peds1-deficient larvae but was mitigated by exogenous plasmalogen, which also alleviated hyper-susceptibility to bacterial infection, as did pharmacological inhibition of caspase-3 and colony-stimulating factor 3-induced myelopoiesis. Overall, our results highlight an important role for plasmalogens in myeloid cell biology and inflammation.
Collapse
Affiliation(s)
- Ana B Arroyo
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Sylwia D Tyrkalska
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Eva Bastida-Martínez
- Departamento de Genética y Microbiología, Área de Genética (Unidad Asociada al IQFR-CSIC), Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
| | - Antonio J Monera-Girona
- Departamento de Genética y Microbiología, Área de Genética (Unidad Asociada al IQFR-CSIC), Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
| | - Joaquín Cantón-Sandoval
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Martín Bernal-Carrión
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain
| | - Diana García-Moreno
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Montserrat Elías-Arnanz
- Departamento de Genética y Microbiología, Área de Genética (Unidad Asociada al IQFR-CSIC), Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
| | - Victoriano Mulero
- Inmunidad, Inflamación y Cáncer. Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain.
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla, 30120, Murcia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
7
|
Li W, Zhang M, Wang Y, Zhao S, Xu P, Cui Z, Chen J, Xia P, Zhang Y. PRRSV GP5 inhibits the antivirus effects of chaperone-mediated autophagy by targeting LAMP2A. mBio 2024; 15:e0053224. [PMID: 38940560 PMCID: PMC11323736 DOI: 10.1128/mbio.00532-24] [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: 02/20/2024] [Accepted: 05/17/2024] [Indexed: 06/29/2024] Open
Abstract
Autophagy is an important biological process in host defense against viral infection. However, many viruses have evolved various strategies to disrupt the host antiviral system. Porcine reproductive and respiratory syndrome virus (PRRSV) is a typical immunosuppressive virus with a large economic impact on the swine industry. At present, studies on the escape mechanism of PRRSV in the autophagy process, especially through chaperone-mediated autophagy (CMA), are limited. This study confirmed that PRRSV glycoprotein 5 (GP5) could disrupt the formation of the GFAP-LAMP2A complex by inhibiting the MTORC2/PHLPP1/GFAP pathway, promoting the dissociation of the pGFAP-EF1α complex, and blocking the K63-linked polyubiquitination of LAMP2A to inhibit the activity of CMA. Further research demonstrated that CMA plays an anti-PRRSV role by antagonizing nonstructural protein 11 (NSP11)-mediated inhibition of type I interferon (IFN-I) signaling. Taken together, these results indicate that PRRSV GP5 inhibits the antiviral effect of CMA by targeting LAMP2A. This research provides new insight into the escape mechanism of immunosuppressive viruses in CMA. IMPORTANCE Viruses have evolved sophisticated mechanisms to manipulate autophagy to evade degradation and immune responses. Porcine reproductive and respiratory syndrome virus (PRRSV) is a typical immunosuppressive virus that causes enormous economic losses in the swine industry. However, the mechanism by which PRRSV manipulates autophagy to defend against host antiviral effects remains unclear. In this study, we found that PRRSV GP5 interacts with LAMP2A and disrupts the formation of the GFAP-LAMP2A complex, thus inhibiting the activity of CMA and subsequently enhancing the inhibitory effect of the NSP11-mediated IFN-I signaling pathway, ultimately facilitating PRRSV replication. Our study revealed a novel mechanism by which PRRSV escapes host antiviral effects through CMA, providing a potential host target, LAMP2A, for developing antiviral drugs and contributing to understanding the escape mechanism of immunosuppressive viruses.
Collapse
Affiliation(s)
- Wen Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Mengting Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yueshuai Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Shijie Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Pengli Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Zhiying Cui
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Jing Chen
- College of Life Science, Henan Agricultural University, Zhengzhou, Henan, China
| | - Pingan Xia
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan, China
| | - Yina Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Chen S, Tie M, Wu M, He A, Chen Y. Blockage of TMEM189 induces G2/M arrest and inhibits the growth of breast tumors. Biochem Biophys Rep 2024; 38:101744. [PMID: 38873225 PMCID: PMC11170349 DOI: 10.1016/j.bbrep.2024.101744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 05/08/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024] Open
Abstract
Cancer is the major cause of premature death in humans worldwide, demanding more efficient therapeutics. Aberrant cell proliferation resulting from the loss of cell cycle regulation is the major hallmark of cancer, so targeting cell cycle is a promising strategy to combat cancer. However, the molecular mechanism underlying the dysregulation of cell cycle of cancer cells remains poorly understood. TMEM189, a newly identified protein, plays roles in the biosynthesis of ethanolamine plasmalogen and the regulation of autophagy. Here, we demonstrated that the expression level of TMEM189 was negatively correlated with the survival rate of the cancer patients. TMEM189 deficiency significantly suppresses the cancer cell proliferation and migration, and causes cell cycle G2/M arrest both in vitro and in vivo. Furthermore, TMEM189 depletion suppressed the growth of breast tumors in vivo. Taken together, our work indicated that TMEM189 promotes cancer progression by regulating cell cycle G2/M transition, suggesting that it is a promising target in cancer therapy.
Collapse
Affiliation(s)
| | | | - Mengyue Wu
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Anyuan He
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Yali Chen
- School of Life Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Papin M, Bouchet AM, Chantôme A, Vandier C. Ether-lipids and cellular signaling: A differential role of alkyl- and alkenyl-ether-lipids? Biochimie 2023; 215:50-59. [PMID: 37678745 DOI: 10.1016/j.biochi.2023.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/17/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Ether-lipids (EL) are specific lipids bearing a characteristic sn-1 ether bond. Depending on the ether or vinyl-ether nature of this bond, they are present as alkyl- or alkenyl-EL, respectively. Among EL, alkenyl-EL, also referred as plasmalogens in the literature, attract most of the scientific interest as they are the predominant EL species in eukaryotic cells, thus less is known about alkyl-EL. EL have been implicated in various signaling pathways and alterations in their quantity are frequently observed in pathologies such as neurodegenerative and cardiovascular diseases or cancer. However, it remains unknown whether both alkyl- and alkenyl-EL play the same roles in these processes. This review summarizes the roles and mechanisms of action of EL in cellular signaling and tries to discriminate between alkyl- and alkenyl-EL. We also focus on the involvement of EL-mediated alterations of cellular signaling in diseases and discuss the potential interest for EL in therapy.
Collapse
Affiliation(s)
- Marion Papin
- Nutrition, Croissance, Cancer (N2C) UMR 1069, University of Tours, INSERM, 37000, Tours, France.
| | | | - Aurélie Chantôme
- Nutrition, Croissance, Cancer (N2C) UMR 1069, University of Tours, INSERM, 37000, Tours, France
| | - Christophe Vandier
- Nutrition, Croissance, Cancer (N2C) UMR 1069, University of Tours, INSERM, 37000, Tours, France; Lifesome Therapeutics, López de Hoyos 42, 28006, Madrid, Spain
| |
Collapse
|
10
|
Cen X, Li Z, Chen X. Ubiquitination in the regulation of autophagy. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1348-1357. [PMID: 37587758 PMCID: PMC10520486 DOI: 10.3724/abbs.2023149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/01/2023] [Indexed: 08/18/2023] Open
Abstract
Autophagy, an efficient and effective approach to clear rapidly damaged organelles, macromolecules, and other harmful cellular components, enables the recycling of nutrient materials and supply of nutrients to maintain cellular homeostasis. Ubiquitination plays an important regulatory role in autophagy. This paper summarizes the most recent progress in ubiquitin modification in various stages of autophagy, including initiation, elongation, and termination. Moreover, this paper shows that ubiquitination is an important way through which selective autophagy achieves substrate specificity. Furthermore, we note the distinction between monoubiquitination and polyubiquitination in the regulation of autophagy. Compared with monoubiquitination, polyubiquitination is a more common strategy to regulate the activity of the autophagy molecular machinery. In addition, the role of ubiquitination in the closure and fusion of autophagosomes warrants further study. This article not only clarifies the regulatory mechanism of autophagy but also contributes to a deeper understanding of the importance of ubiquitination modification.
Collapse
Affiliation(s)
- Xueyan Cen
- Hubei Key laboratory of Edible Wild Plants Conservation & UtilizationHubei Engineering Research Center of Special Wild Vegetables Breeding and Comprehensive Utilization TechnologySchool of Life ScienceHubei Normal UniversityHuangshi435002China
| | - Ziling Li
- Hubei Key laboratory of Edible Wild Plants Conservation & UtilizationHubei Engineering Research Center of Special Wild Vegetables Breeding and Comprehensive Utilization TechnologySchool of Life ScienceHubei Normal UniversityHuangshi435002China
| | - Xinpeng Chen
- Hubei Key laboratory of Edible Wild Plants Conservation & UtilizationHubei Engineering Research Center of Special Wild Vegetables Breeding and Comprehensive Utilization TechnologySchool of Life ScienceHubei Normal UniversityHuangshi435002China
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
| |
Collapse
|
11
|
Autophagy/Mitophagy Regulated by Ubiquitination: A Promising Pathway in Cancer Therapeutics. Cancers (Basel) 2023; 15:cancers15041112. [PMID: 36831455 PMCID: PMC9954143 DOI: 10.3390/cancers15041112] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Autophagy is essential for organismal development, maintenance of energy homeostasis, and quality control of organelles and proteins. As a selective form of autophagy, mitophagy is necessary for effectively eliminating dysfunctional mitochondria. Both autophagy and mitophagy are linked with tumor progression and inhibition. The regulation of mitophagy and autophagy depend upon tumor type and stage. In tumors, mitophagy has dual roles: it removes damaged mitochondria to maintain healthy mitochondria and energy production, which are necessary for tumor growth. In contrast, mitophagy has been shown to inhibit tumor growth by mitigating excessive ROS production, thus preventing mutation and chromosomal instability. Ubiquitination and deubiquitination are important modifications that regulate autophagy. Multiple E3 ubiquitin ligases and DUBs modulate the activity of the autophagy and mitophagy machinery, thereby influencing cancer progression. In this review, we summarize the mechanistic association between cancer development and autophagy/mitophagy activities regulated by the ubiquitin modification of autophagic proteins. In addition, we discuss the function of multiple proteins involved in autophagy/mitophagy in tumors that may represent potential therapeutic targets.
Collapse
|
12
|
Tokareva A, Chagovets V, Starodubtseva N, Rodionov V, Kometova V, Chingin K, Frankevich V. Lipidomic markers of breast cancer malignant tumor histological types. BIOMEDITSINSKAYA KHIMIYA 2022; 68:375-382. [DOI: 10.18097/pbmc20226805375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The molecular profile of a tumor is associated with its histological type and can be used both to study the mechanisms of tumor progression and to diagnose it. In this work, changes in the lipid profile of a malignant breast tumor and the adjacent tissue were studied. The potential possibility of determining the histological type of the tumor by its lipid profile was evaluated. Lipid profiling was performed by reverse-phase chromato-mass-spectrometric analysis the tissue of lipid extract with identification of lipids by characteristic fragments. Potential lipid markers of the histological type of tumor were determined using the Kruskal-Wallis test. Impact of lipid markers was calculated by MetaboAnalyst. Classification models were built by support vector machines with linear kernel and 1-vs-1 architecture. Models were validated by leave-one out cross-validation. Accuracy of models based on microenvironment tissue, were 99% and 75%, accuracy of models, based on tumor tissue, were 90% and 40% for the positive ion mode and negative ion mode respectively. The lipid profile of marginal (adjacent) tissue can be used for identification histological types of breast cancer. Glycerophospholipid metabolism pathway changes were statistically significant in the adjacent tissue and tumor tissue.
Collapse
Affiliation(s)
- A.O. Tokareva
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V.V. Chagovets
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - N.L. Starodubtseva
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V.V. Rodionov
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V.V. Kometova
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - K.S. Chingin
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang, China
| | - V.E. Frankevich
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia; Siberian State Medical University, Tomsk, 634050 Russia
| |
Collapse
|
13
|
Function and regulation of ULK1: From physiology to pathology. Gene 2022; 840:146772. [PMID: 35905845 DOI: 10.1016/j.gene.2022.146772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/03/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022]
Abstract
The expression of ULK1, a core protein of autophagy, is closely related to autophagic activity. Numerous studies have shown that pathological abnormal expression of ULK1 is associated with various human diseases such as neurological disorders, infections, cardiovascular diseases, liver diseases and cancers. In addition, new advances in the regulation of ULK1 have been identified. Furthermore, targeting ULK1 as a therapeutic strategy for diseases is gaining attention as new corresponding activators or inhibitors are being developed. In this review, we describe the structure and regulation of ULK1 as well as the current targeted activators and inhibitors. Moreover, we highlight the pathological disorders of ULK1 expression and its critical role in human diseases.
Collapse
|