1
|
Jing H, Liu W, Qu GP, Niu D, Jin JB. SUMOylation of AL6 regulates seed dormancy and thermoinhibition in Arabidopsis. THE NEW PHYTOLOGIST 2025; 245:1040-1055. [PMID: 39562527 DOI: 10.1111/nph.20270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024]
Abstract
DELAY OF GERMINATION1 (DOG1) is a critical regulator of seed dormancy and seed thermoinhibition. However, how DOG1 expression is regulated by post-translational modifications and how seeds transmit the high-temperature signal to DOG1 remain largely unknown. ALFIN1-like 6/7 (AL6/7) was previously found to repress DOG1 expression during seed imbibition. Here, we found that AL6/7 represses seed dormancy partly by downregulating DOG1 expression. AtSIZ1, a SUMO E3 ligase, interacts with AL6 and mediates its SUMOylation mainly at the lysine 181 residue. SIZ1-mediated SUMOylation of AL6 is required for repression of DOG1 transcription and seed dormancy. SUMOylation of AL6 is required for its association with the DOG1 locus and protects it from ubiquitination and subsequent 26S proteasome-mediated protein degradation. High temperatures decrease SUMOylation levels of AL6, resulting in downregulation of AL6 protein levels and an increase in DOG1 transcription, which consequently causes reduced seed germination. Taken together, these results demonstrate that reversible SUMOylation of AL6 fine-tunes DOG1 expression, which is required for precise establishment of seed dormancy and inhibition of seed germination under high-temperature conditions in Arabidopsis thaliana.
Collapse
Affiliation(s)
- Hua Jing
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- China National Botanical Garden, Beijing, 100093, China
| | - Wei Liu
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- China National Botanical Garden, Beijing, 100093, China
| | - Gao-Ping Qu
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - De Niu
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Bo Jin
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- China National Botanical Garden, Beijing, 100093, China
- Academician Workstation of Agricultural High-tech Industrial Area of the Yellow River Delta, National Center of Technology Innovation for Comprehensive Utilization of Saline-Alkali Land, Dongying, Shandong, 257000, China
| |
Collapse
|
2
|
Lin YW, Lin FY, Lai ZH, Tsai CS, Tsai YT, Huang YS, Liu CW. Porphyromonas gingivalis GroEL accelerates abdominal aortic aneurysm formation by matrix metalloproteinase-2 SUMOylation in vascular smooth muscle cells: A novel finding for the activation of MMP-2. Mol Oral Microbiol 2024. [PMID: 39449503 DOI: 10.1111/omi.12487] [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: 08/12/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Infection is a known cause of abdominal aortic aneurysm (AAA), and matrix metalloproteases-2 (MMP-2) secreted by vascular smooth muscle cells (SMCs) plays a key role in the structural disruption of the middle layer of the arteries during AAA progression. The periodontal pathogen Porphyromonas gingivalis is highly associated with the progression of periodontitis. GroEL protein of periodontal pathogens is an important virulence factor that can invade the body through either the bloodstream or digestive tract and is associated with numerous systemic diseases. Although P. gingivalis aggravates AAA by increasing the expression of MMP-2 in animal studies, the molecular mechanism through which P. gingivalis regulates the expression of MMP-2 is still unknown and requires further investigation. In this study, we first confirmed through animal experiments that P. gingivalis GroEL promotes MMP-2 secretion from vascular SMCs, thereby aggravating Ang II-induced aortic remodeling and AAA formation. In addition, rat vascular SMCs and A7r5 cells were used to investigate the underlying mechanisms in vitro. The results demonstrated that GroEL can promote the interaction between the K639 site of MMP-2 and SUMO-1, leading to MMP-2 SUMOylation, which inhibits the reoccurrence of non-K639-mediated monoubiquitylation. Hence, the monoubiquitylation-mediated lysosomal degradation of MMP-2 is inhibited, consequently promoting MMP-2 stability and production. SUMOylation may facilitate intra-endoplasmic reticulum (ER) and Golgi trafficking of MMP-2, thereby enhancing its transport capacity. In conclusion, this is the first report demonstrating the presence of a novel posttranslational modification, SUMOylation, in the MMP family, suggesting that P. gingivalis GroEL may exacerbate AAA formation by increasing MMP-2 production through SUMOylation in vascular SMCs. This study also provides a novel perspective on the role of SUMOylation in MMP-2-induced systemic diseases.
Collapse
Affiliation(s)
- Yi-Wen Lin
- Institute of Oral Biology, National Yang Ming Chiao Tung University (Taipei Campus), Taipei, Taiwan
| | - Feng-Yen Lin
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ze-Hao Lai
- Institute of Oral Biology, National Yang Ming Chiao Tung University (Taipei Campus), Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Yen-Sung Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chen-Wei Liu
- Department of Basic Medical Science, College of Medicine, University of Arizona, Phoenix, Arizona, USA
| |
Collapse
|
3
|
Wei C. The role of glutathione peroxidase 4 in neuronal ferroptosis and its therapeutic potential in ischemic and hemorrhagic stroke. Brain Res Bull 2024; 217:111065. [PMID: 39243947 DOI: 10.1016/j.brainresbull.2024.111065] [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: 05/09/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Ferroptosis is a type of cell death that depends on iron and is driven by lipid peroxidation, playing a crucial role in neuronal death during stroke. A central element in this process is the inactivation of glutathione peroxidase 4 (GPx4), an antioxidant enzyme that helps maintain redox balance by reducing lipid hydroperoxides. This review examines the critical function of GPx4 in controlling neuronal ferroptosis following ischemic and hemorrhagic stroke. We explore the mechanisms through which GPx4 becomes inactivated in various stroke subtypes. In strokes, excess glutamate depletes glutathione (GSH) and products of hemoglobin breakdown overwhelm GPx4. Studies using genetic models with GPx4 deficiency underscore its vital role in maintaining neuronal survival and function. We also consider new therapeutic approaches to enhance GPx4 activity, including novel small molecule activators, adjustments in GSH metabolism, and selenium supplementation. Additionally, we outline the potential benefits of combining these GPx4-focused strategies with other anti-ferroptotic methods like iron chelation and lipoxygenase inhibition for enhanced neuroprotection. Furthermore, we highlight the significance of understanding the timing of GPx4 inactivation during stroke progression to design effective therapeutic interventions.
Collapse
Affiliation(s)
- Chao Wei
- Feinberg school of medicine, Northwestern University, IL 60611, USA
| |
Collapse
|
4
|
Wei B, Yang F, Yu L, Qiu C. Crosstalk between SUMOylation and other post-translational modifications in breast cancer. Cell Mol Biol Lett 2024; 29:107. [PMID: 39127633 DOI: 10.1186/s11658-024-00624-3] [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: 04/06/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Breast cancer represents the most prevalent tumor type and a foremost cause of mortality among women globally. The complex pathophysiological processes of breast cancer tumorigenesis and progression are regulated by protein post-translational modifications (PTMs), which are triggered by different carcinogenic factors and signaling pathways, with small ubiquitin-like modifier (SUMOylation) emerging as a particularly pivotal player in this context. Recent studies have demonstrated that SUMOylation does not act alone, but interacts with other PTMs, such as phosphorylation, ubiquitination, acetylation, and methylation, thereby leading to the regulation of various pathological activities in breast cancer. This review explores novel and existing mechanisms of crosstalk between SUMOylation and other PTMs. Typically, SUMOylation is regulated by phosphorylation to exert feedback control, while also modulates subsequent ubiquitination, acetylation, or methylation. The crosstalk pairs in promoting or inhibiting breast cancer are protein-specific and site-specific. In mechanism, alterations in amino acid side chain charges, protein conformations, or the occupation of specific sites at specific domains or sites underlie the complex crosstalk. In summary, this review centers on elucidating the crosstalk between SUMOylation and other PTMs in breast cancer oncogenesis and progression and discuss the molecular mechanisms contributing to these interactions, offering insights into their potential applications in facilitating novel treatments for breast cancer.
Collapse
Affiliation(s)
- Bajin Wei
- The Department of Breast Surgery, Key Laboratory of Organ Transplantation, Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Luyang Yu
- MOE Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zijingang Campus, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Cong Qiu
- MOE Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zijingang Campus, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Hao B, Chen K, Zhai L, Liu M, Liu B, Tan M. Substrate and Functional Diversity of Protein Lysine Post-translational Modifications. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae019. [PMID: 38862432 PMCID: PMC12016574 DOI: 10.1093/gpbjnl/qzae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 11/11/2023] [Accepted: 01/08/2024] [Indexed: 06/13/2024]
Abstract
Lysine post-translational modifications (PTMs) are widespread and versatile protein PTMs that are involved in diverse biological processes by regulating the fundamental functions of histone and non-histone proteins. Dysregulation of lysine PTMs is implicated in many diseases, and targeting lysine PTM regulatory factors, including writers, erasers, and readers, has become an effective strategy for disease therapy. The continuing development of mass spectrometry (MS) technologies coupled with antibody-based affinity enrichment technologies greatly promotes the discovery and decoding of PTMs. The global characterization of lysine PTMs is crucial for deciphering the regulatory networks, molecular functions, and mechanisms of action of lysine PTMs. In this review, we focus on lysine PTMs, and provide a summary of the regulatory enzymes of diverse lysine PTMs and the proteomics advances in lysine PTMs by MS technologies. We also discuss the types and biological functions of lysine PTM crosstalks on histone and non-histone proteins and current druggable targets of lysine PTM regulatory factors for disease therapy.
Collapse
Affiliation(s)
- Bingbing Hao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaifeng Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| | - Muyin Liu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| |
Collapse
|
6
|
Wang J, Zhu G. A precise prognostic signature in CTNNB1-mutant hepatocellular carcinoma: Prognosis prediction and precision treatment exploration. Heliyon 2023; 9:e22382. [PMID: 38125518 PMCID: PMC10730442 DOI: 10.1016/j.heliyon.2023.e22382] [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: 02/01/2023] [Revised: 09/27/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023] Open
Abstract
Background CTNNB1 mutates in most hepatocellular carcinoma (HCC) which is the most familiar form of liver cancer with high heterogeneity. It is critical to create a specific prognostication methodology and to investigate additional treatment options for CTNNB1-mutant HCCs. Methods A total of 926 samples in five independent cohorts were enrolled in this study, including 127 CTNNB1-mutant samples and 75 estimated CTNNB1-mutant samples. The prognostic signature was constructed by LASSO-Cox regression and evaluated by bioinformatics analyses. The selection of possible drug targets and agents was produced based on the expression profiles and drug sensitivity data of cancer cell lines in two databases. Results A prognostic signature based on 15 genes categorized the CTNNB1-mutant HCCs into two groups with different risks. Compared to low-risk patients, high-risk patients had significantly inferior prognoses. ROC curve and multivariate analysis also indicated the superior performance of our signature on the prognosis estimation, particularly in CTNNB1-mutant HCCs. Besides, the nomogram was constructed according to the prognostic signature with excellent predictive performance confirmed by the calibration curve. Subsequently, we suggested that AT-7519 and PHA-793887 might be potential drug agents for high-risk patients. Conclusion We established a 15-gene prognostic model, particularly in HCCs with CTNNB1 mutations with good predictive efficiency. Besides, we explored the potential drug targets and agents for patients with high risk. Our findings offered a fresh idea for personalized prognosis management in HCCs with CTNNB1 mutations and threw new insight for precise treatment in HCCs as well.
Collapse
Affiliation(s)
- Junying Wang
- Department of Interventional and Vascular Surgery, Zhongda Hospital, Southeast University, Jiangsu, 210009, China
| | - Guangyu Zhu
- Department of Interventional and Vascular Surgery, Zhongda Hospital, Southeast University, Jiangsu, 210009, China
| |
Collapse
|
7
|
Li K, Xia Y, He J, Wang J, Li J, Ye M, Jin X. The SUMOylation and ubiquitination crosstalk in cancer. J Cancer Res Clin Oncol 2023; 149:16123-16146. [PMID: 37640846 DOI: 10.1007/s00432-023-05310-z] [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: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The cancer occurrence and progression are largely affected by the post-translational modifications (PTMs) of proteins. Currently, it has been shown that the relationship between ubiquitination and SUMOylation is highly complex and interactive. SUMOylation affects the process of ubiquitination and degradation of substrates. Contrarily, SUMOylation-related proteins are also regulated by the ubiquitination process thus altering their protein levels or activity. Emerging evidence suggests that the abnormal regulation between this crosstalk may lead to tumorigenesis. PURPOSE In this review, we have discussed the study of the relationship between ubiquitination and SUMOylation, as well as the possibility of a corresponding application in tumor therapy. METHODS The relevant literatures from PubMed have been reviewed for this article. CONCLUSION The interaction between ubiquitination and SUMOylation is crucial for the occurrence and development of cancer. A greater understanding of the crosstalk of SUMOylation and ubiquitination may be more conducive to the development of more selective and effective SUMOylation inhibitors, as well as a promotion of synergy with other tumor treatment strategies.
Collapse
Affiliation(s)
- Kailang Li
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yongming Xia
- Department of Oncology, Yuyao People's Hospital of Zhejiang, Yuyao, 315400, Zhejiang, China
| | - Jian He
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jie Wang
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jingyun Li
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Meng Ye
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China.
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, 315020, China.
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
8
|
Wang K, Papadopoulos N, Hamidi A, Lennartsson J, Heldin CH. SUMOylation of PDGF receptor α affects signaling via PLCγ and STAT3, and cell proliferation. BMC Mol Cell Biol 2023; 24:19. [PMID: 37193980 DOI: 10.1186/s12860-023-00481-6] [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: 12/29/2022] [Accepted: 05/05/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The platelet-derived growth factor (PDGF) family of ligands exerts their cellular effects by binding to α- and β-tyrosine kinase receptors (PDGFRα and PDGFRβ, respectively). SUMOylation is an important posttranslational modification (PTM) which regulates protein stability, localization, activation and protein interactions. A mass spectrometry screen has demonstrated SUMOylation of PDGFRα. However, the functional role of SUMOylation of PDGFRα has remained unknown. RESULTS In the present study, we validated that PDGFRα is SUMOylated on lysine residue 917 as was previously reported using a mass spectrometry approach. Mutation of lysine residue 917 to arginine (K917R) in PDGFRα substantially decreased SUMOylation, indicating that this amino acid residue is a major SUMOylation site. Whereas no difference in the stability of wild-type and mutant receptor was observed, the K917R mutant PDGFRα was less ubiquitinated than wild-type PDGFRα. The internalization and trafficking of the receptor to early and late endosomes were not affected by the mutation, neither was the localization of the PDGFRα to Golgi. However, the K917R mutant PDGFRα showed delayed activation of PLC-γ and enhanced activation of STAT3. Functional assays showed that the mutation of K917 of PDGFRα decreased cell proliferation in response to PDGF-BB stimulation. CONCLUSIONS SUMOylation of PDGFRα decreases ubiquitination of the receptor and affects ligand-induced signaling and cell proliferation.
Collapse
Affiliation(s)
- Kehuan Wang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Box 582, Sweden
| | - Natalia Papadopoulos
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Box 582, Sweden
| | - Anahita Hamidi
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Box 582, Sweden
| | - Johan Lennartsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Box 582, Sweden.
| |
Collapse
|
9
|
Patton BK, Madadi S, Briley SM, Ahmed AA, Pangas SA. Sumoylation regulates functional properties of the oocyte transcription factors SOHLH1 and NOBOX. FASEB J 2023; 37:e22747. [PMID: 36607631 PMCID: PMC10129296 DOI: 10.1096/fj.202201481r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
SOHLH1 and NOBOX are oocyte-expressed transcription factors with critical roles in ovary development and fertility. In mice, Sohlh1 and Nobox are essential for fertility through their regulation of the oocyte transcriptional network and cross-talk to somatic cells. Sumoylation is a posttranslational modification that regulates transcription factor function, and we previously showed that mouse oocytes deficient for sumoylation had an altered transcriptional landscape that included significant changes in NOBOX target genes. Here, we show that mouse SOHLH1 is modified by SUMO2/3 at lysine 345 and mutation of this residue alters SOHLH1 nuclear to cytoplasmic localization. In NOBOX, we identify a non-consensus SUMO site, K97, that eliminates NOBOX mono-SUMO2/3 conjugation, while a point mutation at K125 had no effect on NOBOX sumoylation. However, NOBOXK97R/K125R double mutants showed loss of mono-SUMO2/3 and altered higher molecular weight modifications, suggesting cooperation between these lysine's. NOBOXK97R and NOBOXK97R/K125R differentially regulated NOBOX promoter targets, with increased activity on the Gdf9 promoter, but no effect on the Pou5f1 promoter. These data implicate sumoylation as a novel regulatory mechanism for SOHLH1 and NOBOX, which may prove useful in refining their roles during oogenesis as well as their function during reprogramming to generate de novo germ cells.
Collapse
Affiliation(s)
- Bethany K. Patton
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Surabhi Madadi
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
- Rice University, Houston, TX 77005
| | - Shawn M. Briley
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Avery A. Ahmed
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030
| | - Stephanie A. Pangas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
10
|
Wang M, Wei R, Li G, Bi HL, Jia Z, Zhang M, Pang M, Li X, Ma L, Tang Y. SUMOylation of SYNJ2BP-COX16 promotes breast cancer progression through DRP1-mediated mitochondrial fission. Cancer Lett 2022; 547:215871. [PMID: 35998797 DOI: 10.1016/j.canlet.2022.215871] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022]
Abstract
Treatments targeting oncogenic fusion proteins are notable examples of successful drug development. Abnormal splicing of genes resulting in fusion proteins is a critical driver of various tumors, but the underlying mechanism remains poorly understood. Here, we show that SUMOylation of the fusion protein Synaptojanin 2 binding protein-Cytochrome-c oxidase 16 (SYNJ2BP-COX16) at K107 induces mitochondrial fission in breast cancer and that the K107 site regulates SYNJ2BP-COX16 mitochondrial subcellular localization. Compared with a non-SUMOylated K107R mutant, wild-type SYNJ2BP-COX16 contributed to breast cancer cell proliferation and metastasis in vivo and in vitro by increasing adenosine triphosphate (ATP) production and cytochrome-c oxidase (COX) activity. SUMOylated SYNJ2BP-COX16 recruits dynamin-related protein 1 (DRP1) to the mitochondria to promote ubiquitin-conjugating enzyme 9 (UBC9) binding to DRP1, enhance SUMOylation of DRP1 and phosphorylation of DRP1 at S616, and then induce mitochondrial fission. Moreover, Mdivi-1, an inhibitor of DRP1 phosphorylation, decreased the localization of DRP1 in mitochondria, and prevents SYNJ2BP-COX16 induced mitochondrial fission, cell proliferation and metastasis. Based on these data, SYNJ2BP-COX16 promotes breast cancer progression through the phosphorylation of DRP1 and subsequent induction of mitochondrial fission, indicating that SUMOylation at the K107 residue of SYNJ2BP-COX16 is a novel potential treatment target for breast cancer.
Collapse
Affiliation(s)
- Miao Wang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Ranru Wei
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Guohui Li
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China; College of New Materials and Chemical Engineering, Beijing Key Laboratory of Enze Biomass Fine Chemicals, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116024, China.
| | - Zhaojun Jia
- College of New Materials and Chemical Engineering, Beijing Key Laboratory of Enze Biomass Fine Chemicals, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Mengjie Zhang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Mengyao Pang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Xiaona Li
- School of Environmental Science and Technology, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Liming Ma
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Ying Tang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| |
Collapse
|
11
|
Cui C, Yang F, Li Q. Post-Translational Modification of GPX4 is a Promising Target for Treating Ferroptosis-Related Diseases. Front Mol Biosci 2022; 9:901565. [PMID: 35647032 PMCID: PMC9133406 DOI: 10.3389/fmolb.2022.901565] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/28/2022] [Indexed: 12/23/2022] Open
Abstract
Glutathione peroxidase 4 (GPX4) is one of the most important antioxidant enzymes. As the key regulator of ferroptosis, GPX4 has attracted considerable attention in the fields of cancer, cardiovascular, and neuroscience research in the past 10 years. How to regulate GPX4 activity has become a hot topic nowadays. GPX4 protein level is regulated transcriptionally by transcription factor SP2 or Nrf2. GPX4 activity can be upregulated by supplementing intracellular selenium or glutathione, and also be inhibited by ferroptosis inducers such as ML162 and RSL3. These regulatory mechanisms of GPX4 level/activity have already shown a great potential for treating ferroptosis-related diseases in preclinical studies, especially in cancer cells. Until recently, research show that GPX4 can undergo post-translational modifications (PTMs), such as ubiquitination, succination, phosphorylation, and glycosylation. PTMs of GPX4 affect the protein level/activity of GPX4, indicating that modifying these processes can be a potential therapy for treating ferroptosis-related diseases. This article summarizes the protein characteristics, enzyme properties, and PTMs of GPX4. It also provides a hypothetical idea for treating ferroptosis-related diseases by targeting the PTMs of GPX4.
Collapse
Affiliation(s)
- Can Cui
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qian Li
- Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
- *Correspondence: Qian Li,
| |
Collapse
|
12
|
Li YZ, Zhang C, Pei JP, Zhang WC, Zhang CD, Dai DQ. The functional role of Pescadillo ribosomal biogenesis factor 1 in cancer. J Cancer 2022; 13:268-277. [PMID: 34976188 PMCID: PMC8692700 DOI: 10.7150/jca.58982] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Tumors are neogrowths formed by the growth of normal cells or tissues through complex mechanisms under the influence of many factors. The occurrence and development of tumors are affected by many factors. Pescadillo ribosomal biogenesis factor 1 (PES1) has been identified as a cancer-related gene. The study of these genes may open up new avenues for early diagnosis, treatment and prognosis of tumors. As a nucleolar protein and part of the Pes1/Bop1/WDR12 (PeBoW) complex, PES1 is involved in ribosome biogenesis and DNA replication. Many studies have shown that high expression of PES1 is often closely related to the occurrence, proliferation, invasion, metastasis, prognosis and sensitivity to chemotherapeutics of various human malignant tumors through a series of molecular mechanisms and signaling pathways. The molecules that regulate the expression of PES1 include microRNA (miRNA), circular RNA (circRNA), c-Jun, bromodomain-containing protein 4 (BRD4) and nucleolar phosphoprotein B23. However, the detailed pathogenic mechanisms of PES1 overexpression in human malignancies remains unclear. This article summarizes the role of PES1 in the carcinogenesis, prognosis and treatment of multiple tumors, and introduces the molecular mechanisms and signal transduction pathways related to PES1.
Collapse
Affiliation(s)
- Yong-Zhi Li
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Cheng Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Jun-Peng Pei
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Wan-Chuan Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Chun-Dong Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.,Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Dong-Qiu Dai
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.,Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
13
|
Bian Z, Zhou M, Cui K, Yang F, Cao Y, Sun S, Liu B, Gong L, Li J, Wang X, Li C, Yao S, Yin Y, Huang S, Fei B, Huang Z. SNHG17 promotes colorectal tumorigenesis and metastasis via regulating Trim23-PES1 axis and miR-339-5p-FOSL2-SNHG17 positive feedback loop. J Exp Clin Cancer Res 2021; 40:360. [PMID: 34782005 PMCID: PMC8591805 DOI: 10.1186/s13046-021-02162-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/30/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Small nucleolar RNA host gene (SNHG) long noncoding RNAs (lncRNAs) are frequently dysregulated in human cancers and involved in tumorigenesis and progression. SNHG17 has been reported as a candidate oncogene in several cancer types, however, its regulatory role in colorectal cancer (CRC) is unclear. METHODS SNHG17 expression in multiple CRC cohorts was assessed by RT-qPCR or bioinformatic analyses. Cell viability was evaluated using Cell Counting Kit-8 (CCK-8) and colony formation assays. Cell mobility and invasiveness were assessed by Transwell assays. Tumor xenograft and metastasis models were applied to confirm the effects of SNHG17 on CRC tumorigenesis and metastasis in vivo. Immunohistochemistry staining was used to measure protein expression in cancer tissues. RNA pull-down, RNA immunoprecipitation, chromatin immunoprecipitation, and dual luciferase assays were used to investigate the molecular mechanism of SNHG17 in CRC. RESULTS Using multiple cohorts, we confirmed that SNHG17 is aberrantly upregulated in CRC and correlated with poor survival. In vitro and in vivo functional assays indicated that SNHG17 facilitates CRC proliferation and metastasis. SNHG17 impedes PES1 degradation by inhibiting Trim23-mediated ubiquitination of PES1. SNHG17 upregulates FOSL2 by sponging miR-339-5p, and FOSL2 transcription activates SNHG17 expression, uncovering a SNHG17-miR-339-5p-FOSL2-SNHG17 positive feedback loop. CONCLUSIONS We identified SNHG17 as an oncogenic lncRNA in CRC and identified abnormal upregulation of SNHG17 as a prognostic risk factor for CRC. Our mechanistic investigations demonstrated, for the first time, that SNHG17 promotes tumor growth and metastasis through two different regulatory mechanisms, SNHG17-Trim23-PES1 axis and SNHG17-miR-339-5p-FOSL2-SNHG17 positive feedback loop, which may be exploited for CRC therapy.
Collapse
Affiliation(s)
- Zehua Bian
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Mingyue Zhou
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Fan Yang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Shengbai Sun
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Bingxin Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Liang Gong
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jiuming Li
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Xue Wang
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Chaoqun Li
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Surui Yao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Bojian Fei
- Department of Surgical Oncology, Affiliated Hospital of Jiangnan University, 200 Hui He Road, Wuxi, 214062, Jiangsu, China.
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
14
|
Qin Y, Yuan H, Chen X, Yang X, Xing Z, Shen Y, Dong W, An S, Qi Y, Wu H. SUMOylation Wrestles With the Occurrence and Development of Breast Cancer. Front Oncol 2021; 11:659661. [PMID: 33968766 PMCID: PMC8097099 DOI: 10.3389/fonc.2021.659661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer has the highest incidence among cancers and is the most frequent cause of death in women worldwide. The detailed mechanism of the pathogenesis of breast cancer has not been fully elucidated, and there remains a lack of effective treatment methods for the disease. SUMOylation covalently conjugates a large amount of cellular proteins, and affects their cellular localization and biological activity to participate in numerous cellular processes. SUMOylation is an important process and imbalance of SUMOylation results in the progression of human diseases. Increasing evidence shows that numerous SUMOylated proteins are involved in the occurrence and development of breast cancer. This review summarizes a series of studies on protein SUMOylation in breast cancer in recent years. The study of SUMOylated proteins provides a comprehensive understanding of the pathophysiology of breast cancer and provides evolving therapeutic strategies for the treatment of breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yitao Qi
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Hongmei Wu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
15
|
Liu S, Wang L, Jiang D, Wei W, Nasir MF, Khan MS, Yousafi Q, Liu X, Fu X, Li X, Li J. Sumoylation as an Emerging Target in Therapeutics against Cancer. Curr Pharm Des 2021; 26:4764-4776. [PMID: 32568016 DOI: 10.2174/1381612826666200622124134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Sumoylation is the Post-translational modification gaining most of the research interest recently. Sumoylation is involved in various crucial functions of the cell such as regulation of cell cycle, DNA damage repair, apoptosis, etc. Oncology is advancing in radiotherapy, targeted chemotherapy, various forms of immunotherapy and targeted gene therapy. Researches are being conducted to prove its connotation with a variety of cancers and inhibitors are being developed to obstruct the fatal effect caused by misbalance of the SUMO-catalytic cycle. It has been shown that up-regulation of certain enzymes of Sumoylation correlates with cancer incidence in most of the cases. However, in some cases, down-regulation also associates with cancer invasion such as underexpression of UBC9 in initial stage breast cancer. This can aid in future study, treatment, and diagnosis of a variety of cancers including breast cancer, prostate cancer, lung adenocarcinoma, melanoma, multiple myeloma, etc. Various mechanistic assays are being developed and used to identify potential inhibitors against the dysregulated proteins of Sumoylation. This review summarizes the normal roles of the enzymes involved in the SUMOcatalytic cycle, their misbalanced regulation leading to tumorigenesis and nearly all the potent inhibitors identified to date, while after detailed studied it was observed that ML-792 could be a promising inhibitor in treating cancers by inhibiting Sumoylation enzymes.
Collapse
Affiliation(s)
- Sitong Liu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, Jilin, China,College of Life Sciences, Jilin University, Changchun, 130012, China
| | - Lichun Wang
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, Jilin, China
| | - Dongjun Jiang
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, Jilin, China
| | - Wei Wei
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, Jilin, China,Dental Hospital, Jilin University, Changchun 130021, China
| | - Mushyeda Fatima Nasir
- Department of Biosciences, Faculty of Sciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Muhammad Saad Khan
- Department of Biosciences, Faculty of Sciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Qudsia Yousafi
- Department of Biosciences, Faculty of Sciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Xintong Liu
- Dental Hospital, Jilin University, Changchun 130021, China
| | - Xueqi Fu
- College of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, Jilin, China
| | - Jiang Li
- Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China,Dental Hospital, Jilin University, Changchun 130021, China
| |
Collapse
|
16
|
Fu Z, Jiao Y, Li YQ, Ke JJ, Xu YH, Jia BX, Liu B. PES1 In Liver Cancer: A Prognostic Biomarker With Tumorigenic Roles. Cancer Manag Res 2019; 11:9641-9653. [PMID: 31814761 PMCID: PMC6861535 DOI: 10.2147/cmar.s226471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Liver cancer has a high incidence of mortality. DNA replication and posttranscriptional modifications play important roles in the development of liver cancer. Pescadillo (PES1) is a nuclear protein that is involved in embryonic development, ribosome synthesis, DNA replication, and cell cycle progression. Recently, abnormal PES1 expression was reported in several tumors, including neuroblastoma, colon cancer, gastric cancer, and breast cancer. Based on bio-informatic analysis, cell experiments and animal models, the aim of this study is to investigate the expression patterns and specific roles of PES1 in liver cancer. PATIENTS AND METHODS PES1 expression was represented by boxplots. The correlation between PES1 expression and clinical features was assessed by the chi-squared test and Fisher's exact tests. Kaplan-Meier curves compared overall survival between different levels of PES1 expression, and Cox analysis selected potential variables associated with overall survival. The MTT assay investigated the proliferation rate, the scratch assay assessed the migratory ability, and the Transwell assay evaluated the invasion capacity of tumor cells in vitro. Animal models were used to confirm the tumorigenic roles of PES1 in vivo. GSEA illustrated the molecular mechanisms that PES1 participated in. RESULTS We found that PES1 was highly expressed in liver cancer tissues, served as a diagnostic marker, and correlated with poor overall survival (OS) and relapse-free survival (RFS) in patients. In vitro studies indicated that PES1 promoted tumor cell proliferation (P=0.0034), migration (P=0.0026), and invasion (P=0.0008), and this tumorigenic role was confirmed in animal models. GSEA further illuminated molecular mechanisms that PES1 participated in liver cancer occurrence and progression. CONCLUSION This study suggested that PES1 was upregulated in liver cancer and correlated with poor prognosis, by promoting tumor cell proliferation, migration, and invasion, and PES1 may be a novel diagnostic and prognostic bio-marker and a promising therapeutic target in liver cancer.
Collapse
Affiliation(s)
- Zhuo Fu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Yan-Qing Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Jian-Ji Ke
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Yan-Hui Xu
- Department of Digestive, China-Japan Union Hospital Affiliated to Jilin University, Changchun, Jilin130033, People’s Republic of China
| | - Bao-Xing Jia
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| |
Collapse
|
17
|
Wu Z, Huang R, Yuan L. Crosstalk of intracellular post-translational modifications in cancer. Arch Biochem Biophys 2019; 676:108138. [PMID: 31606391 DOI: 10.1016/j.abb.2019.108138] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/29/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022]
Abstract
Post-translational modifications (PTMs) have been reported to play pivotal roles in numerous cellular biochemical and physiological processes. Multiple PTMs can influence the actions of each other positively or negatively, termed as PTM crosstalk or PTM code. During recent years, development of identification strategies for PTMs co-occurrence has revealed abundant information of interplay between PTMs. Increasing evidence demonstrates that deregulation of PTMs crosstalk is involved in the genesis and development of various diseases. Insight into the complexity of PTMs crosstalk will help us better understand etiology and provide novel targets for drug therapy. In the present review, we will discuss the important functional roles of PTMs crosstalk in proteins associated with cancer diseases.
Collapse
Affiliation(s)
- Zheng Wu
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, 100191, China.
| | - Rongting Huang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Liang Yuan
- Peking University International Hospital, Beijing, 102200, China
| |
Collapse
|
18
|
Ma L, Herren AW, Espinal G, Randol J, McLaughlin B, Martinez-Cerdeño V, Pessah IN, Hagerman RJ, Hagerman PJ. Composition of the Intranuclear Inclusions of Fragile X-associated Tremor/Ataxia Syndrome. Acta Neuropathol Commun 2019; 7:143. [PMID: 31481131 PMCID: PMC6720097 DOI: 10.1186/s40478-019-0796-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/24/2019] [Indexed: 12/11/2022] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder associated with a premutation repeat expansion (55-200 CGG repeats) in the 5' noncoding region of the FMR1 gene. Solitary intranuclear inclusions within FXTAS neurons and astrocytes constitute a hallmark of the disorder, yet our understanding of how and why these bodies form is limited. Here, we have discovered that FXTAS inclusions emit a distinct autofluorescence spectrum, which forms the basis of a novel, unbiased method for isolating FXTAS inclusions by preparative fluorescence-activated cell sorting (FACS). Using a combination of autofluorescence-based FACS and liquid chromatography/tandem mass spectrometry (LC-MS/MS)-based proteomics, we have identified more than two hundred proteins that are enriched within the inclusions relative to FXTAS whole nuclei. Whereas no single protein species dominates inclusion composition, highly enriched levels of conjugated small ubiquitin-related modifier 2 (SUMO 2) protein and p62/sequestosome-1 (p62/SQSTM1) protein were found within the inclusions. Many additional proteins involved with RNA binding, protein turnover, and DNA damage repair were enriched within inclusions relative to total nuclear protein. The current analysis has also allowed the first direct detection, through peptide sequencing, of endogenous FMRpolyG peptide, the product of repeat-associated non-ATG (RAN) translation of the FMR1 mRNA. However, this peptide was found only at extremely low levels and not within whole FXTAS nuclear preparations, raising the question whether endogenous RAN products exist at quantities sufficient to contribute to FXTAS pathogenesis. The abundance of the inclusion-associated ubiquitin- and SUMO-based modifiers supports a model for inclusion formation as the result of increased protein loads and elevated oxidative stress leading to maladaptive autophagy. These results highlight the need to further investigate FXTAS pathogenesis in the context of endogenous systems.
Collapse
Affiliation(s)
- Lisa Ma
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, One Shields Ave, Davis, CA, USA
| | - Anthony W Herren
- Genome Center, University of California Davis, Davis, California, USA
| | - Glenda Espinal
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, One Shields Ave, Davis, CA, USA
| | - Jamie Randol
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, One Shields Ave, Davis, CA, USA
| | - Bridget McLaughlin
- Department of Pathology and Laboratory Medicine, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Veronica Martinez-Cerdeño
- Department of Pathology and Laboratory Medicine, University of California Davis, School of Medicine, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospital of Northern California, University of California Davis, School of Medicine, Sacramento, California, USA
- MIND Institute, University of California Davis Health, Sacramento, California, USA
| | - Isaac N Pessah
- MIND Institute, University of California Davis Health, Sacramento, California, USA
- Department of Molecular Biosciences, University of California Davis, School of Veterinary Medicine, Davis, California, USA
| | - Randi J Hagerman
- MIND Institute, University of California Davis Health, Sacramento, California, USA
- Department of Pediatrics, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Paul J Hagerman
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, One Shields Ave, Davis, CA, USA.
- MIND Institute, University of California Davis Health, Sacramento, California, USA.
| |
Collapse
|
19
|
Wei S, Liu K, He Q, Gao Y, Shen L. PES1 is regulated by CD44 in liver cancer stem cells via miR-105-5p. FEBS Lett 2019; 593:1777-1786. [PMID: 31127852 DOI: 10.1002/1873-3468.13459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
Pescadillo (PES1) is a key molecule for ribosome formation in mammalian cells. In this study, human hepatoma C3A cells were reprogrammed by four transcription factors, Oct4, Sox2, Klf4 and c-Myc, into induced cancer stem cells, termed C3A-induced cancer stem cells (C3A-iCSCs). We found that PES1 was up-regulated in C3A-iCSCs and promoted cell proliferation. Moreover, the cancer stem cell marker CD44, which is located in the cytomembrane, translocated to the nucleus and was up-regulated in C3A-iCSCs. Our results suggest that CD44 has a negative effect on miR-105-5p. We found that PES1 is a direct target of, and was negatively regulated by, miR-105-5p. In summary, CD44 regulates PES1 in liver cancer stem cells via miR-105-5p to promote cell growth.
Collapse
Affiliation(s)
- Shiruo Wei
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Haidian District, Beijing, China
| | - Kaiyu Liu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qihua He
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Haidian District, Beijing, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Li Shen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Haidian District, Beijing, China
| |
Collapse
|
20
|
Yang Y, He Y, Wang X, Liang Z, He G, Zhang P, Zhu H, Xu N, Liang S. Protein SUMOylation modification and its associations with disease. Open Biol 2018; 7:rsob.170167. [PMID: 29021212 PMCID: PMC5666083 DOI: 10.1098/rsob.170167] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/31/2017] [Indexed: 02/05/2023] Open
Abstract
SUMOylation, as a post-translational modification, plays essential roles in various biological functions including cell growth, migration, cellular responses to stress and tumorigenesis. The imbalance of SUMOylation and deSUMOylation has been associated with the occurrence and progression of various diseases. Herein, we summarize and discuss the signal crosstalk between SUMOylation and ubiquitination of proteins, protein SUMOylation relations with several diseases, and the identification approaches for SUMOylation site. With the continuous development of bioinformatics and mass spectrometry, several accurate and high-throughput methods have been implemented to explore small ubiquitin-like modifier-modified substrates and sites, which is helpful for deciphering protein SUMOylation-mediated molecular mechanisms of disease.
Collapse
Affiliation(s)
- Yanfang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China
| | - Yu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China
| | - Xixi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China
| | - Ziwei Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China
| | - Gu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China
| | - Peng Zhang
- Department of Urinary Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100034, People's Republic of China
| | - Ningzhi Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100034, People's Republic of China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, People's Republic of China
| |
Collapse
|
21
|
Xu Z, Yang Y, Li B, Li Y, Xia K, Yang Y, Li X, Wang M, Li S, Wu H. Checkpoint suppressor 1 suppresses transcriptional activity of ERα and breast cancer cell proliferation via deacetylase SIRT1. Cell Death Dis 2018; 9:559. [PMID: 29752474 PMCID: PMC5948204 DOI: 10.1038/s41419-018-0629-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 02/08/2023]
Abstract
Breast cancer is a highly heterogeneous carcinoma in women worldwide, but the underlying mechanisms that account for breast cancer initiation and development have not been fully established. Mounting evidence indicates that Checkpoint suppressor 1 (CHES1) is tightly associated with tumorigenesis and prognosis in many types of cancer. However, the definitive function of CHES1 in breast cancer remains to be explored. Here we showed that CHES1 had a physical interaction with estrogen receptor-α (ERα) and repressed the transactivation of ERα in breast cancer cells. Mechanistically, the interaction between CHES1 and ERα enhanced the recruitment of nicotinamide adenine dinucleotide (NAD+) deacetylase Sirtuin 1 (SIRT1), and it further induced SIRT1-mediated ERα deacetylation and repression on the promoter-binding enrichment of ERα. In addition, we also found that the expression of CHES1 was repressed by estrogen-ERα signaling and the expression level of CHES1 was significantly downregulated in ERα-positive breast cancer. The detailed mechanism was that ERα may directly bind to CHES1 potential promoter via recognizing the conserved estrogen response element (ERE) motif in response to estrogen stimulation. Functionally, CHES1 inhibited ERα-mediated proliferation and tumorigenesis of breast cancer cells in vivo and in vitro. Totally, these results identified a negative cross-regulatory loop between ERα and CHES1 that was required for growth of breast cancer cells, it might uncover novel insight into molecular mechanism of CHES1 involved in breast cancer and provide new avenues for molecular-targeted therapy in hormone-regulated breast cancer.
Collapse
Affiliation(s)
- Zhaowei Xu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yangyang Yang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Bowen Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yanan Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Kangkai Xia
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yuxi Yang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Xiahui Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Miao Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Shujing Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Huijian Wu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China.
| |
Collapse
|
22
|
Chen H, Xu Z, Li X, Yang Y, Li B, Li Y, Xia K, Wang J, Li S, Wang M, Wu H. α-catenin SUMOylation increases IκBα stability and inhibits breast cancer progression. Oncogenesis 2018. [PMID: 29540699 PMCID: PMC5852976 DOI: 10.1038/s41389-018-0037-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
α-catenin has been demonstrated to suppress several different types of cancers. Here we demonstrate that α-catenin is modified by SUMO protein, which covalently binds α-catenin at the carboxy terminus at lysine 870. Substitution of lysine 870 with arginine completely abolishes α-catenin SUMOylation. This modification can be removed by SENP1. However, α-catenin SUMOylation does not affect its stability and subcellular localization. In addition, we observed that the SUMOylation-deficient α-catenin mutant has a reduced interaction with IκBα which prevents subsequent ubiquitination of IκBα, and therefore a reduced suppression of expression of the NF-κB target genes TNF-α, IL-8, VEGF, and uPA. In addition, the α-catenin SUMOylation mutant shows impaired suppression of tumor growth. These results demonstrate that SUMOylation at lysine 870 of α-catenin plays a key role in the suppression of the NF-κB pathway, which inhibits breast cancer tumor growth and migration.
Collapse
Affiliation(s)
- Huan Chen
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Zhaowei Xu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Xiahui Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yangyang Yang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Bowen Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yanan Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Kangkai Xia
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Jian Wang
- School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Shujing Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Miao Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China.
| | - Huijian Wu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China. .,School of Life Science and Medicine, Dalian University of Technology, Panjin, China.
| |
Collapse
|
23
|
Han ZJ, Feng YH, Gu BH, Li YM, Chen H. The post-translational modification, SUMOylation, and cancer (Review). Int J Oncol 2018; 52:1081-1094. [PMID: 29484374 PMCID: PMC5843405 DOI: 10.3892/ijo.2018.4280] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/14/2018] [Indexed: 02/07/2023] Open
Abstract
SUMOylation is a reversible post-translational modification which has emerged as a crucial molecular regulatory mechanism, involved in the regulation of DNA damage repair, immune responses, carcinogenesis, cell cycle progression and apoptosis. Four SUMO isoforms have been identified, which are SUMO1, SUMO2/3 and SUMO4. The small ubiquitin-like modifier (SUMO) pathway is conserved in all eukaryotes and plays pivotal roles in the regulation of gene expression, cellular signaling and the maintenance of genomic integrity. The SUMO catalytic cycle includes maturation, activation, conjugation, ligation and de-modification. The dysregulation of the SUMO system is associated with a number of diseases, particularly cancer. SUMOylation is widely involved in carcinogenesis, DNA damage response, cancer cell proliferation, metastasis and apoptosis. SUMO can be used as a potential therapeutic target for cancer. In this review, we briefly outline the basic concepts of the SUMO system and summarize the involvement of SUMO proteins in cancer cells in order to better understand the role of SUMO in human disease.
Collapse
Affiliation(s)
- Zhi-Jian Han
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yan-Hu Feng
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Bao-Hong Gu
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yu-Min Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Hao Chen
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
24
|
Pfister AS, Kühl M. Of Wnts and Ribosomes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:131-155. [PMID: 29389514 DOI: 10.1016/bs.pmbts.2017.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Wnt proteins are secreted glycoproteins that activate different intracellular signal transduction pathways. They regulate cell proliferation and are required for proper embryonic development. Misregulation of Wnt signaling can result in various diseases including cancer. In most circumstances, cell growth is essential for cell division and thus cell proliferation. Therefore, several reports have highlighted the key role of Wnt proteins for cell growth. Ribosomes represent the cellular protein synthesis machinery and cells need to be equipped with an appropriate number of ribosomes to allow cell growth. Recent findings suggest a role for Wnt proteins in regulating ribosome biogenesis and we here summarize these findings representing a previously unknown function of Wnt proteins. Understanding this role of Wnt signaling might open new avenues to slow down proliferation by drugs for instance in cancer therapy.
Collapse
Affiliation(s)
- Astrid S Pfister
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany.
| | - Michael Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| |
Collapse
|
25
|
Wang M, Wu H, Li S, Xu Z, Li X, Yang Y, Li B, Li Y, Guo J, Chen H. SYNJ2BP promotes the degradation of PTEN through the lysosome-pathway and enhances breast tumor metastasis via PI3K/AKT/SNAI1 signaling. Oncotarget 2017; 8:89692-89706. [PMID: 29163781 PMCID: PMC5685702 DOI: 10.18632/oncotarget.21058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/03/2017] [Indexed: 12/22/2022] Open
Abstract
SYNJ2BP plays an important role in breast cancer metastasis. However, the molecular mechanism associated with the function of SYNJ2BP in metastasis remains unclear. In this study, we investigated the role of SYNJ2BP in tumor metastasis and established the associated underlying mechanism. Over-expression of SYNJ2BP promoted both cell migration and invasion. In contrast, silencing SYNJ2BP caused the suppression of cell migration and invasion. SYNJ2BP increased the levels of phosphorylation for AKT and GSK3β, which could be inhibited by the PI3K inhibitor, LY294002, and the GSK3β inhibitor, LiCl, and regulated the accumulation of SNAI1 in the nucleus and the expression of the SNAI1 target gene, E-cadherin (EMT marker). It is known that the stability of PTEN is regulated by ubiquitination. However, in this study, we additionally demonstrated that SYNJ2BP mediated the degradation of PTEN protein by the lysosome-pathway and induced the activation of PI3K/AKT signaling by promoting the co-localization of PTEN with autophagy-lysosomes and the expression of LC3-II and p62. In vivo study, the overexpression of SYNJ2BP significantly increased the metastasis of 4T1 cells in BALB/c mice. In addition, SYNJ2BP was highly expressed in breast carcinoma (p = 0.0031), but not in normal breast tissue, while analysis of tissue samples taken from SNAI1-positive human breast cancers showed a significant correlation between the expression of SYNJ2BP and that of p-AKT (p < 0.005). Collectively, our data identified a tumor inducer, SYNJ2BP, which could activate the PI3K/AKT/GSK3β/SNAI1 signaling pathway through the lysosome-mediated degradation of PTEN, and promote both EMT and tumor metastasis during the progression of breast cancer.
Collapse
Affiliation(s)
- Miao Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Huijian Wu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China.,School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Shujing Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Zhaowei Xu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Xiahui Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yangyang Yang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Bowen Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yanan Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Jing Guo
- School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Huan Chen
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| |
Collapse
|