1
|
Zhang C, Wang D, Shen Y, Zhang Y, Liu J. Nuclear translocation of CDK5RAP3 regulated by NXF3 promotes the progression of gastric cancer. Cell Mol Life Sci 2025; 82:100. [PMID: 40032765 PMCID: PMC11876495 DOI: 10.1007/s00018-025-05630-y] [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: 09/04/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Nuclear-cytoplasmic transport proteins (NCTPs) impact the transport of proteins and RNA molecules between the nucleus and cytoplasm in tumor cells, making them promising targets for cancer therapy. Currently, the molecular mechanism and function of Nuclear RNA export factor 3 (NXF3) in gastric cancer (GC) remains unclear. METHODS We used Univariate Cox regression analysis and LASSO regression analysis, Receiver Operating Characteristic (ROC) curves to construct and evaluate a NCTP prognosis risk scoring model (NCTP model). Moreover, we identified the key NCTP (NXF3) affecting GC through differential expression and prognosis analysis. Subsequently, we introduced NXF3 shRNA into GC cells to investigate the impact of NXF3 on the cell proliferation, cell migration, invasion, and cell cycle and apoptosis and tumor growth by CCK-8 assay, transwell, wound healing assay, Flow cytometry, and nude mice subcutaneous tumor in vitro and in vivo. Furthermore, we investigated the key molecules influenced by NXF3 through piRNA-Seq, RNA-Seq, RIP-Seq, IP-MS, and Nuclear-cytoplasmic transcriptomics. RESULTS We constructed a prognostic risk model related to 3 NCTPs, including NXF3, GLE1 and RANGAP. The NCTP model effectively predicts the prognosis of GC patients. The low-risk group exhibited a significantly higher overall survival rate than that of the high-risk group. Notably, NXF3 is identified as a crucial NCTP in GC, and its high expression is associated with poor prognosis of GC patients. Knocking down of NXF3 significantly inhibited the proliferation, invasion, migration, cell cycle, tumor growth and induced cell apoptosis of GC cells in vitro and in vivo. Mechanistically, NXF3 modulates the cell cycle, cellular senescence related oncogenic pathways via piRNA-target network. Specifically, our findings highlighted several piRNA-related signaling pathways in GC, such as piRNA_3457319-CCND1/CDKN1A-p53, piRNA_2847077-TGFB3/TGFBR2-Cellular senescence, piRNA_448895-IGF1/PDGFRA/ACTB/MAP2K6-Rap1. Moreover, NXF3 was shown to facilitate the nuclear export of CDK5RAP3 mRNA, thereby promoting cell cycle progression and increasing cancer cell proliferation in gastric cancer. CONCLUSION Our study demonstrates that NXF3 modulates cell cycle progression and promotes gastric cancer development through piRNA-related pathways and the nuclear export of CDK5RAP3 mRNA. Targeting NXF3 represents a promising strategy for developing novel therapeutic approaches for gastric cancer.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Dongyang Wang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Yuguang Shen
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Yuanruohan Zhang
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China
| | - Jiahua Liu
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated, Shanghai Jiaotong University School of Medicine, No.160, Pujian Road, Pudong New Area, Shanghai, 200127, China.
| |
Collapse
|
2
|
Zhu G, Tong N, Zhu Y, Wang L, Wang Q. The crosstalk between SUMOylation and immune system in host-pathogen interactions. Crit Rev Microbiol 2025; 51:164-186. [PMID: 38619159 DOI: 10.1080/1040841x.2024.2339259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/16/2024]
Abstract
Pathogens can not only cause infectious diseases, immune system diseases, and chronic diseases, but also serve as potential triggers or initiators for certain tumors. They directly or indirectly damage human health and are one of the leading causes of global deaths. Small ubiquitin-like modifier (SUMO) modification, a type of protein post-translational modification (PTM) that occurs when SUMO groups bond covalently to particular lysine residues on substrate proteins, plays a crucial role in both innate and adaptive immunologic responses, as well as pathogen-host immune system crosstalk. SUMOylation participates in the host's defense against pathogens by regulating immune responses, while numerically vast and taxonomically diverse pathogens have evolved to exploit the cellular SUMO modification system to break through innate defenses. Here, we describe the characteristics and multiple functions of SUMOylation as a pivotal PTM mechanism, the tactics employed by various pathogens to counteract the immune system through targeting host SUMOylation, and the character of the SUMOylation system in the fight between pathogens and the host immune system. We have also included a summary of the potential anti-pathogen SUMO enzyme inhibitors. This review serves as a reference for basic research and clinical practice in the diagnosis, prognosis, and treatment of pathogenic microorganism-caused disorders.
Collapse
Affiliation(s)
- Gangli Zhu
- Guangdong Province Solid Waste Recycling and Heavy Metal Pollution Control Engineering Technology Research Center, Guangdong Polytechnic of Environment Protection Engineering, Foshan, Guangdong, China
| | - Ni Tong
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yipeng Zhu
- Guagnzhou NO.6 Middle school, Guangzhou, Guangdong, China
| | - Lize Wang
- General Department, Institute of Software Chinese Academy of Sciences, Beijing, China
| | - Qirui Wang
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Zhang X, Hu Q, Peng H, Huang J, Sang W, Guan J, Huang Z, Jiang B, Sun D. Therapeutic potential of flavopiridol in diabetic retinopathy: Targeting DDX58. Int Immunopharmacol 2024; 137:112504. [PMID: 38897127 DOI: 10.1016/j.intimp.2024.112504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/02/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Diabetic retinopathy (DR), a common complication of diabetes, is characterized by inflammation and neovascularization, and is intricately regulated by the ubiquitin-proteasome system (UPS). Despite advancements, identifying ubiquitin-related genes and drugs specifically targeting DR remains a significant challenge. In this study, bioinformatics analyses and the Connectivity Map (CMAP) database were utilized to explore the therapeutic potential of genes and drugs for DR. Through these methodologies, flavopiridol was identified as a promising therapeutic candidate. To evaluate flavopiridol's therapeutic potential in DR, an in vitro model using Human Umbilical Vein Endothelial Cells (HUVECs) induced by high glucose (HG) conditions was established. Additionally, in vivo models using mice with streptozotocin (STZ)-induced DR and oxygen-induced retinopathy (OIR) were employed. The current study reveals that flavopiridol possesses robust anti-inflammatory and anti-neovascularization properties. To further elucidate the molecular mechanisms of flavopiridol, experimental validation and molecular docking techniques were employed. These efforts identified DDX58 as a predictive target for flavopiridol. Notably, our research demonstrated that flavopiridol modulates the DDX58/NLRP3 signaling pathway, thereby exerting its therapeutic effects in suppressing inflammation and neovascularization in DR. This study unveils groundbreaking therapeutic agents and innovative targets for DR, and establishes a progressive theoretical framework for the application of ubiquitin-related therapies in DR.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Qiang Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Hongsong Peng
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Jiayang Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Wei Sang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Jitian Guan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Zhangxin Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Bo Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Dawei Sun
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
4
|
Li X, Yang P, Hou X, Ji S. Post-Translational Modification of PTEN Protein: Quantity and Activity. Oncol Rev 2024; 18:1430237. [PMID: 39144161 PMCID: PMC11321960 DOI: 10.3389/or.2024.1430237] [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: 05/09/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024] Open
Abstract
Post-translational modifications play crucial roles in regulating protein functions and stabilities. PTEN is a critical tumor suppressor involved in regulating cellular proliferation, survival, and migration processes. However, dysregulation of PTEN is common in various human cancers. PTEN stability and activation/suppression have been extensively studied in the context of tumorigenesis through inhibition of the PI3K/AKT signaling pathway. PTEN undergoes various post-translational modifications, primarily including phosphorylation, acetylation, ubiquitination, SUMOylation, neddylation, and oxidation, which finely tune its activity and stability. Generally, phosphorylation modulates PTEN activity through its lipid phosphatase function, leading to altered power of the signaling pathways. Acetylation influences PTEN protein stability and degradation rate. SUMOylation has been implicated in PTEN localization and interactions with other proteins, affecting its overall function. Neddylation, as a novel modification of PTEN, is a key regulatory mechanism in the loss of tumor suppressor function of PTEN. Although current therapeutic approaches focus primarily on inhibiting PI3 kinase, understanding the post-translational modifications of PTEN could help provide new therapeutic strategies that can restore PTEN's role in PIP3-dependent tumors. The present review summarizes the major recent developments in the regulation of PTEN protein level and activity. We expect that these insights will contribute to better understanding of this critical tumor suppressor and its potential implications for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiao Li
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Pu Yang
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Xiaoli Hou
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Shaoping Ji
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng, Henan, China
| |
Collapse
|
5
|
Gou Y, Liu D, Chen M, Wei Y, Huang X, Han C, Feng Z, Zhang C, Lu T, Peng D, Xue Y. GPS-SUMO 2.0: an updated online service for the prediction of SUMOylation sites and SUMO-interacting motifs. Nucleic Acids Res 2024; 52:W238-W247. [PMID: 38709873 PMCID: PMC11223847 DOI: 10.1093/nar/gkae346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
Small ubiquitin-like modifiers (SUMOs) are tiny but important protein regulators involved in orchestrating a broad spectrum of biological processes, either by covalently modifying protein substrates or by noncovalently interacting with other proteins. Here, we report an updated server, GPS-SUMO 2.0, for the prediction of SUMOylation sites and SUMO-interacting motifs (SIMs). For predictor training, we adopted three machine learning algorithms, penalized logistic regression (PLR), a deep neural network (DNN), and a transformer, and used 52 404 nonredundant SUMOylation sites in 8262 proteins and 163 SIMs in 102 proteins. To further increase the accuracy of predicting SUMOylation sites, a pretraining model was first constructed using 145 545 protein lysine modification sites, followed by transfer learning to fine-tune the model. GPS-SUMO 2.0 exhibited greater accuracy in predicting SUMOylation sites than did other existing tools. For users, one or multiple protein sequences or identifiers can be input, and the prediction results are shown in a tabular list. In addition to the basic statistics, we integrated knowledge from 35 public resources to annotate SUMOylation sites or SIMs. The GPS-SUMO 2.0 server is freely available at https://sumo.biocuckoo.cn/. We believe that GPS-SUMO 2.0 can serve as a useful tool for further analysis of SUMOylation and SUMO interactions.
Collapse
Affiliation(s)
- Yujie Gou
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Dan Liu
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Miaomiao Chen
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Yuxiang Wei
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Xinhe Huang
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Cheng Han
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Zihao Feng
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Chi Zhang
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Teng Lu
- Computer Network Information Center, Chinese Academy of Sciences, Beijing100190, China
| | - Di Peng
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
| | - Yu Xue
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan430074, China
- Nanjing University Institute of Artificial Intelligence Biomedicine, Nanjing210031, China
| |
Collapse
|
6
|
Madahar V, Dang R, Zhang Q, Liu C, Rodgers VGJ, Liao J. Human Post-Translational SUMOylation Modification of SARS-CoV-2 Nucleocapsid Protein Enhances Its Interaction Affinity with Itself and Plays a Critical Role in Its Nuclear Translocation. Viruses 2023; 15:1600. [PMID: 37515286 PMCID: PMC10384427 DOI: 10.3390/v15071600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/25/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Viruses, such as Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), infect hosts and take advantage of host cellular machinery for genome replication and new virion production. Identifying and elucidating host pathways for viral infection is critical for understanding the development of the viral life cycle and novel therapeutics. The SARS-CoV-2 N protein is critical for viral RNA (vRNA) genome packaging in new virion formation. Using our quantitative Förster energy transfer/Mass spectrometry (qFRET/MS) coupled method and immunofluorescence imaging, we identified three SUMOylation sites of the SARS-CoV-2 N protein. We found that (1) Small Ubiquitin-like modifier (SUMO) modification in Nucleocapsid (N) protein interaction affinity increased, leading to enhanced oligomerization of the N protein; (2) one of the identified SUMOylation sites, K65, is critical for its nuclear translocation. These results suggest that the host human SUMOylation pathway may be critical for N protein functions in viral replication and pathology in vivo. Thus, blocking essential host pathways could provide a novel strategy for future anti-viral therapeutics development, such as for SARS-CoV-2 and other viruses.
Collapse
Affiliation(s)
- Vipul Madahar
- Department of Bioengineering, College of Engineering, Bourns College of Engineering, University of California at Riverside, Riverside, CA 92521, USA
| | - Runrui Dang
- Department of Bioengineering, College of Engineering, Bourns College of Engineering, University of California at Riverside, Riverside, CA 92521, USA
| | - Quanqing Zhang
- Institute for Integrative Genome Biology, University of California at Riverside, Riverside, CA 92521, USA
- Department of Botany, College of Natural & Agricultural Sciences, University of California at Riverside, Riverside, CA 92521, USA
| | - Chuchu Liu
- Department of Bioengineering, College of Engineering, Bourns College of Engineering, University of California at Riverside, Riverside, CA 92521, USA
| | - Victor G J Rodgers
- Department of Bioengineering, College of Engineering, Bourns College of Engineering, University of California at Riverside, Riverside, CA 92521, USA
- Biomedical Science, School of Medicine, University of California at Riverside, Riverside, CA 92521, USA
| | - Jiayu Liao
- Department of Bioengineering, College of Engineering, Bourns College of Engineering, University of California at Riverside, Riverside, CA 92521, USA
- Institute for Integrative Genome Biology, University of California at Riverside, Riverside, CA 92521, USA
- Biomedical Science, School of Medicine, University of California at Riverside, Riverside, CA 92521, USA
| |
Collapse
|
7
|
Huang J, Tan X, Liu Y, Jiang K, Luo J. Knockdown of UBE2I inhibits tumorigenesis and enhances chemosensitivity of cholangiocarcinoma via modulating p27kip1 nuclear export. Mol Carcinog 2023; 62:700-715. [PMID: 36825757 DOI: 10.1002/mc.23518] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/25/2023]
Abstract
The asymptomatic nature of cholangiocarcinoma (CCA), particularly during its early stages, in combination with its high aggressiveness and chemoresistance, significantly compromises the efficacy of current therapeutic options, contributing to a dismal prognosis. As a tumor suppressor that inhibits the cell cycle, abnormal cytoplasmic p27kip1 localization is related to chemotherapy resistance and often occurs in various cancers, including CCA. Nevertheless, the underlying mechanism is unclear. SUMOylation, which is involved in regulating subcellular localization and the cell cycle, is a posttranslational modification that regulates p27kip1 activity. Here, we confirmed that UBE2I, as the only key enzyme for SUMOylation, was highly expressed and p27kip1 was downregulated in CCA tissues, which were associated with poor outcomes in CCA. Moreover, UBE2I silencing inhibited CCA cell proliferation, delayed xenograft tumor growth in vivo, and sensitized CCA cells to the chemotherapeutics, which may be due to cell cycle arrest induced by p27kip1 nuclear accumulation. According to the immunoprecipitation result, we found that UBE2I could bind p27kip1, and the binding amount of p27kip1 and SUMO-1 decreased after UBE2I silencing. Moreover, nuclear retention of p27kip1 was induced by UBE2I knockdown and SUMOylation or CRM1 inhibition, further suggesting that UBE2I could cooperate with CRM1 in the nuclear export of p27kip1. These data indicate that UBE2I-mediated SUMOylation is a novel regulatory mechanism that underlies p27kip1 export and controls CCA tumorigenesis, providing a therapeutic option for CCA treatment.
Collapse
Affiliation(s)
- Jie Huang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiaolong Tan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan Liu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Kainian Jiang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jian Luo
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|
8
|
SENP6-Mediated deSUMOylation of VEGFR2 Enhances Its Cell Membrane Transport in Angiogenesis. Int J Mol Sci 2023; 24:ijms24032544. [PMID: 36768878 PMCID: PMC9916989 DOI: 10.3390/ijms24032544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
Angiogenesis is a significant pathogenic characteristic of diabetic microangiopathy. Advanced glycation end products (AGEs) are considerably elevated in diabetic tissues and can affect vascular endothelial cell shape and function. Regulation of the vascular endothelial growth factor (VEGF)-VEGF receptor 2 (VEGFR2) signaling pathway is a critical mechanism in the regulation of angiogenesis, and VEGFR2 activity can be modified by post-translational changes. However, little research has been conducted on the control of small ubiquitin-related modifier (SUMO)-mediated VEGFR2 alterations. The current study investigated this using human umbilical vein endothelial cells (HUVECs) in conjunction with immunoblotting and immunofluorescence. AGEs increased Nrf2 translocation to the nucleus and promoted VEGFR2 expression. They also increased the expression of sentrin/SUMO-specific protease 6 (SENP6), which de-SUMOylated VEGFR2, and immunofluorescence indicated a reduction in VEGFR2 accumulation in the Golgi and increased VEGFR2 transport from the Golgi to the cell membrane surface via the coatomer protein complex subunit beta 2. VEGFR2 on the cell membrane was linked to VEGF generated by pericytes, triggering the VEGF signaling cascade. In conclusion, this study demonstrates that SENP6 regulates VEGFR2 trafficking from the Golgi to the endothelial cell surface. The SENP6-VEGFR2 pathway plays a critical role in pathological angiogenesis.
Collapse
|
9
|
Chen S, Liu Q, Zhang L, Ma J, Xue B, Li H, Deng R, Guo M, Xu Y, Tian R, Wang J, Cao W, Yang Q, Wang L, Li X, Liu S, Yang D, Zhu H. The Role of REC8 in the Innate Immune Response to Viral Infection. J Virol 2022; 96:e0217521. [PMID: 35107381 PMCID: PMC8941933 DOI: 10.1128/jvi.02175-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/20/2022] Open
Abstract
REC8 meiotic recombination protein (REC8) is a member of structural maintenance of chromosome (SMC) protein partners, which play an important role in meiosis, antitumor activity, and sperm formation. As the adaptor proteins of RIG-I-like receptor (RLR) signaling and cyclic GMP-AMP synthase (cGAS)-DNA signaling, the activity and stability of MAVS (mitochondrial antiviral signaling protein; also known as VISA, Cardif, and IPS-1) and STING (stimulator of interferon genes; also known as MITA) are critical for innate immunity. Here, we report that REC8 interacts with MAVS and STING and inhibits their ubiquitination and subsequent degradation, thereby promoting innate antiviral signaling. REC8 is upregulated through the JAK-STAT signaling pathway during viral infection. Knockdown of REC8 impairs the innate immune responses against vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), and herpes simplex virus (HSV). Mechanistically, during infection with viruses, the SUMOylated REC8 is transferred from the nucleus to the cytoplasm and then interacts with MAVS and STING to inhibit their K48-linked ubiquitination triggered by RNF5. Moreover, REC8 promotes the recruitment of TBK1 to MAVS and STING. Thus, REC8 functions as a positive modulator of innate immunity. Our work highlights a previously undocumented role of meiosis-associated protein REC8 in regulating innate immunity. IMPORTANCE The innate immune response is crucial for the host to resist the invasion of viruses and other pathogens. STING and MAVS play a critical role in the innate immune response to DNA and RNA viral infection, respectively. In this study, REC8 promoted the innate immune response by targeting STING and MAVS. Notably, REC8 interacts with MAVS and STING in the cytoplasm and inhibits K48-linked ubiquitination of MAVS and STING triggered by RNF5, stabilizing MAVS and STING protein to promote innate immunity and gradually inhibiting viral infection. Our study provides a new insight for the study of antiviral innate immunity.
Collapse
Affiliation(s)
- Shengwen Chen
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Qian Liu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Lini Zhang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Jiahuan Ma
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Binbin Xue
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Huiyi Li
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Rilin Deng
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Mengmeng Guo
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Yan Xu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Renyun Tian
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Jingjing Wang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Wenyan Cao
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Qiong Yang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Luolin Wang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Xinran Li
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Shun Liu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Di Yang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Haizhen Zhu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
- Research Center of Cancer Prevention & Treatment, Translational Medicine Research Center of Liver Cancer, Hunan Cancer Hospital, Changsha, China
| |
Collapse
|
10
|
Hua D, Wu X. Small-molecule inhibitors targeting small ubiquitin-like modifier pathway for the treatment of cancers and other diseases. Eur J Med Chem 2022; 233:114227. [PMID: 35247754 DOI: 10.1016/j.ejmech.2022.114227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/12/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023]
Abstract
SUMOylation is a key post-translational modification that involves the covalent attachment of small ubiquitin-like modifier (SUMO) to the lysine residues of target proteins. The well-balanced SUMOylation is essential for normal cellular behaviors, while disturbance of SUMOylation is associated with various cancers and other diseases. Herein, we summarize the structures and biological functions of proteins involved in the SUMOylation process, their dysregulation in human diseases, and the discovery of small-molecular inhibitors targeting this pathway. In addition, we highlight the emerging trends in this field.
Collapse
Affiliation(s)
- Dexiang Hua
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoxing Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
11
|
Main Factors Involved in Thyroid Hormone Action. Molecules 2021; 26:molecules26237337. [PMID: 34885918 PMCID: PMC8658769 DOI: 10.3390/molecules26237337] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022] Open
Abstract
The thyroid hormone receptors are the mediators of a multitude of actions by the thyroid hormones in cells. Most thyroid hormone activities require interaction with nuclear receptors to bind DNA and regulate the expression of target genes. In addition to genomic regulation, thyroid hormones function via activation of specific cytosolic pathways, bypassing interaction with nuclear DNA. In the present work, we reviewed the most recent literature on the characteristics and roles of different factors involved in thyroid hormone function in particular, we discuss the genomic activity of thyroid hormone receptors in the nucleus and the functions of different thyroid hormone receptor isoforms in the cytosol. Furthermore, we describe the integrin αvβ3-mediated thyroid hormone signaling pathway and its rapid nongenomic action in the cell. We furthermore reviewed the thyroid hormone transporters enabling the uptake of thyroid hormones in the cell, and we also include a paragraph on the proteins that mediate thyroid receptors’ shuttling from the nucleus to the cytosol.
Collapse
|
12
|
Abstract
SUMOylation is a reversible posttranslational modification involved in the regulation of diverse biological processes. Growing evidence suggests that virus infection can interfere with the SUMOylation system. In the present study, we discovered that apoptosis inhibitor 5 (API5) is a SUMOylated protein. Amino acid substitution further identified that Lys404 of API5 was the critical residue for SUMO3 conjugation. Moreover, we found that Avibirnavirus infectious bursal disease virus (IBDV) infection significantly decreased SUMOylation of API5. In addition, our results further revealed that viral protein VP3 inhibited the SUMOylation of API5 by targeting API5 and promoting UBC9 proteasome-dependent degradation through binding to the ubiquitin E3 ligase TRAF3. Furthermore, we revealed that wild-type but not K404R mutant API5 inhibited IBDV replication by enhancing MDA5-dependent IFN-β production. Taken together, our data demonstrate that API5 is a UBC9-dependent SUMOylated protein and deSUMOylation of API5 by viral protein VP3 aids in viral replication.
Collapse
|
13
|
Kim IG, Lee JH, Kim SY, Heo CK, Kim RK, Cho EW. Targeting therapy-resistant lung cancer stem cells via disruption of the AKT/TSPYL5/PTEN positive-feedback loop. Commun Biol 2021; 4:778. [PMID: 34163000 PMCID: PMC8222406 DOI: 10.1038/s42003-021-02303-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer stem cells (CSCs) are regarded as essential targets to overcome tumor progression and therapeutic resistance; however, practical targeting approaches are limited. Here, we identify testis-specific Y-like protein 5 (TSPYL5) as an upstream regulator of CSC-associated genes in non-small cell lung cancer cells, and suggest as a therapeutic target for CSC elimination. TSPYL5 elevation is driven by AKT-dependent TSPYL5 phosphorylation at threonine-120 and stabilization via inhibiting its ubiquitination. TSPYL5-pT120 also induces nuclear translocation and functions as a transcriptional activator of CSC-associated genes, ALDH1 and CD44. Also, nuclear TSPYL5 suppresses the transcription of PTEN, a negative regulator of PI3K signaling. TSPYL5-pT120 maintains persistent CSC-like characteristics via transcriptional activation of CSC-associated genes and a positive feedback loop consisting of AKT/TSPYL5/PTEN signaling pathway. Accordingly, elimination of TSPYL5 by inhibiting TSPYL5-pT120 can block aberrant AKT/TSPYL5/PTEN cyclic signaling and TSPYL5-mediated cancer stemness regulation. Our study suggests TSPYL5 be an effective target for therapy-resistant cancer. In order to assist the development of cancer stem cell (CSC) therapy, Kim et al identified testis-specific Y-like protein 5 (TSPYL5) as an upstream regulator of CSC-associated genes in non-small cell lung cancer cells. They demonstrated in cancer cell lines and in vivo that TSPYL5 activity is dependent on AKT signalling and that disruption of TSPYL5 signalling could serve as a potential strategy to tackle therapy-resistant cancers.
Collapse
Affiliation(s)
- In-Gyu Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon, South Korea. .,Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon, South Korea.
| | - Jei-Ha Lee
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon, South Korea
| | - Seo-Yeon Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon, South Korea
| | - Chang-Kyu Heo
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Rae-Kwon Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon, South Korea.,Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon, South Korea
| | - Eun-Wie Cho
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.
| |
Collapse
|
14
|
Martin JG, Ward JA, Feyertag F, Zhang L, Couvertier S, Guckian K, Huber KVM, Johnson DS. Chemoproteomic Profiling of Covalent XPO1 Inhibitors to Assess Target Engagement and Selectivity. Chembiochem 2021; 22:2116-2123. [PMID: 33887086 DOI: 10.1002/cbic.202100038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/19/2021] [Indexed: 11/11/2022]
Abstract
Selinexor, a covalent XPO1 inhibitor, is approved in the USA in combination with dexamethasone for penta-refractory multiple myeloma. Additional XPO1 covalent inhibitors are currently in clinical trials for multiple diseases including hematologic malignancies, solid tumor malignancies, glioblastoma multiforme (GBM), and amyotrophic lateral sclerosis (ALS). It is important to measure the target engagement and selectivity of covalent inhibitors to understand the degree of engagement needed for efficacy, while avoiding both mechanism-based and off-target toxicity. Herein, we report clickable probes based on the XPO1 inhibitors selinexor and eltanexor for the labeling of XPO1 in live cells to assess target engagement and selectivity. We used mass spectrometry-based chemoproteomic workflows to profile the proteome-wide selectivity of selinexor and eltanexor and show that they are highly selective for XPO1. Thermal profiling analysis of selinexor further offers an orthogonal approach to measure XPO1 engagement in live cells. We believe these probes and assays will serve as useful tools to further interrogate the biology of XPO1 and its inhibition in cellular and in vivo systems.
Collapse
Affiliation(s)
- Jeffrey G Martin
- Biogen, Chemical Biology & Proteomics 225 Binney Street, Cambridge, MA 02142, USA
| | - Jennifer A Ward
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Felix Feyertag
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Lu Zhang
- Biogen, Chemical Biology & Proteomics 225 Binney Street, Cambridge, MA 02142, USA
| | - Shalise Couvertier
- Biogen, Chemical Biology & Proteomics 225 Binney Street, Cambridge, MA 02142, USA
| | - Kevin Guckian
- Biogen, Medicinal Chemistry 225 Binney Street, Cambridge, MA 02142, USA
| | - Kilian V M Huber
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Douglas S Johnson
- Biogen, Chemical Biology & Proteomics 225 Binney Street, Cambridge, MA 02142, USA
| |
Collapse
|
15
|
Anyetei-Anum CS, Evans RM, Back AM, Roggero VR, Allison LA. Acetylation modulates thyroid hormone receptor intracellular localization and intranuclear mobility. Mol Cell Endocrinol 2019; 495:110509. [PMID: 31319097 PMCID: PMC6708479 DOI: 10.1016/j.mce.2019.110509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/05/2019] [Accepted: 07/14/2019] [Indexed: 01/07/2023]
Abstract
The thyroid hormone receptor (TR) undergoes nucleocytoplasmic shuttling, but is primarily nuclear-localized and mediates expression of genes involved in development and homeostasis. Given the proximity of TR acetylation and sumoylation sites to nuclear localization (NLS) and nuclear export signals, we investigated their role in regulating intracellular localization. The nuclear/cytosolic fluorescence ratio (N/C) of fluorescent protein-tagged acetylation mimic, nonacetylation mimic, and sumoylation-deficient TR was quantified in transfected mammalian cells. While nonacetylation mimic and sumoylation-deficient TRs displayed wild-type N/C, the acetylation mimic's N/C was significantly lower. Importins that interact with wild-type TR also interact with acetylation and nonacetylation mimics, suggesting factors other than reduced importin binding alter nuclear localization. FRAP analysis showed wild-type intranuclear dynamics of acetylation mimic and sumoylation-deficient TRs, whereas the nonacetylation mimic had significantly reduced mobility and transcriptional activity. Acetyltransferase CBP/p300 inhibition enhanced TR's nuclear localization, further suggesting that nonacetylation correlates with nuclear retention, while acetylation promotes cytosolic localization.
Collapse
Affiliation(s)
- Cyril S Anyetei-Anum
- Department of Biology, College of William and Mary, 540 Landrum Drive, Integrated Science Center 3030, Williamsburg, VA, 23187, USA
| | - Rochelle M Evans
- Department of Biology, College of William and Mary, 540 Landrum Drive, Integrated Science Center 3030, Williamsburg, VA, 23187, USA
| | - Amanda M Back
- Department of Biology, College of William and Mary, 540 Landrum Drive, Integrated Science Center 3030, Williamsburg, VA, 23187, USA
| | - Vincent R Roggero
- Department of Biology, College of William and Mary, 540 Landrum Drive, Integrated Science Center 3030, Williamsburg, VA, 23187, USA
| | - Lizabeth A Allison
- Department of Biology, College of William and Mary, 540 Landrum Drive, Integrated Science Center 3030, Williamsburg, VA, 23187, USA.
| |
Collapse
|
16
|
Sabbir MG. Progesterone induced Warburg effect in HEK293 cells is associated with post-translational modifications and proteasomal degradation of progesterone receptor membrane component 1. J Steroid Biochem Mol Biol 2019; 191:105376. [PMID: 31067491 DOI: 10.1016/j.jsbmb.2019.105376] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/17/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
Progesterone (P4) is a major steroid hormone that has important effects on metabolism. The progesterone receptor membrane component 1 (PGRMC1) is a non-canonical P4 binding protein. The biological functions affected by PGRMC1 include cholesterol/steroid biosynthesis and metabolism, iron homeostasis and heme trafficking, autophagy, regulation of cell cycle and proliferation, cell migration and invasion. PGRMC1 has been an attractive target for therapeutic intervention in cancer and neurodegenerative disorders due to its biological role in promoting cell survival. P4 has been used in a number of clinical applications and is considered neuroprotective. The involvement of PGRMC1 in P4-mediated regulation of cellular glucose metabolism is not well studied. PGRMC1 is a 21 kDa protein but complex post-translational modifications (PTMs) lead to the existence of several high molecular mass proteins whose molecular function, intracellular distribution, and physiological relevancies are not fully known. Therefore, in this study, P4-PGRMC1-mediated cellular glucose metabolism and PTMs of PGRMC1 were studied using wild-type and CRISPR/Cas9 mediated PGRMC1 knockout (KO) human embryonic kidney-derived (HEK293) cell lines. A 70 kDa (p70) and 100 kDa (p100) PGRMC1 proteins were identified that are predominantly associated with endoplasmic reticulum/mitochondria and nuclear fractions in the cells, respectively. Phosphorylation, acetylation, ubiquitination, and sumoylation of native PGRMC1 under serum starvation were identified which provided an explanation for the higher molecular masses. This study indicates that P4-PGRMC1 signaling caused a rapid increase in glycolysis in the presence of oxygen (aerobic glycolysis) and a corresponding decrease in cellular respiration, known as the Warburg effect. Further, it was demonstrated that the P4-induced increase in glycolysis is associated with rapid proteasomal degradation of the p70 and reduction of the nuclear p100 protein level. P4 treatment also caused significant alteration in the dynamics of PGRMC1 PTMs and its association with potential interacting proteins. Overall, this study provides a hitherto unknown aspect of P4-PGRMC1 mediated signaling that changes basic cellular metabolism in HEK293 cells.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, R2H 2A6, Canada.
| |
Collapse
|
17
|
Bennetzen MV, Kosar M, Bunkenborg J, Payne MR, Bartkova J, Lindström MS, Lukas J, Andersen JS, Bartek J, Larsen DH. DNA damage-induced dynamic changes in abundance and cytosol-nuclear translocation of proteins involved in translational processes, metabolism, and autophagy. Cell Cycle 2018; 17:2146-2163. [PMID: 30196736 DOI: 10.1080/15384101.2018.1515552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Ionizing radiation (IR) causes DNA double-strand breaks (DSBs) and activates a versatile cellular response regulating DNA repair, cell-cycle progression, transcription, DNA replication and other processes. In recent years proteomics has emerged as a powerful tool deepening our understanding of this multifaceted response. In this study we use SILAC-based proteomics to specifically investigate dynamic changes in cytoplasmic protein abundance after ionizing radiation; we present in-depth bioinformatics analysis and show that levels of proteins involved in autophagy (cathepsins and other lysosomal proteins), proteasomal degradation (Ubiquitin-related proteins), energy metabolism (mitochondrial proteins) and particularly translation (ribosomal proteins and translation factors) are regulated after cellular exposure to ionizing radiation. Downregulation of no less than 68 ribosomal proteins shows rapid changes in the translation pattern after IR. Additionally, we provide evidence of compartmental cytosol-nuclear translocation of numerous DNA damage related proteins using protein correlation profiling. In conclusion, these results highlight unexpected cytoplasmic processes actively orchestrated after genotoxic insults and protein translocation from the cytoplasm to the nucleus as a fundamental regulatory mechanism employed to aid cell survival and preservation of genome integrity.
Collapse
Affiliation(s)
- Martin V Bennetzen
- a Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology , University of Southern Denmark , Odense M , Denmark
| | - Martin Kosar
- b Genome Integrity Unit, Danish Cancer Society Research Center , Danish Cancer Society , Copenhagen , Denmark
| | - Jakob Bunkenborg
- a Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology , University of Southern Denmark , Odense M , Denmark
| | - Mark Ronald Payne
- c National Institute of Aquatic Resources , Technical University of Denmark , Lyngby , Denmark
| | - Jirina Bartkova
- b Genome Integrity Unit, Danish Cancer Society Research Center , Danish Cancer Society , Copenhagen , Denmark.,d Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Genome Biology , Karolinska Institutet , Solna , Sweden
| | - Mikael S Lindström
- d Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Genome Biology , Karolinska Institutet , Solna , Sweden
| | - Jiri Lukas
- e Protein Signaling Program, The Novo Nordisk Foundation Center for Protein Research , Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens S Andersen
- a Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology , University of Southern Denmark , Odense M , Denmark
| | - Jiri Bartek
- b Genome Integrity Unit, Danish Cancer Society Research Center , Danish Cancer Society , Copenhagen , Denmark.,d Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Genome Biology , Karolinska Institutet , Solna , Sweden
| | - Dorthe Helena Larsen
- b Genome Integrity Unit, Danish Cancer Society Research Center , Danish Cancer Society , Copenhagen , Denmark.,f Nucleolar Stress and Disease Group, Danish Cancer Society Research Center , Danish Cancer Society , Copenhagen , Denmark
| |
Collapse
|
18
|
Abstract
Pancreatic cancer is an aggressive and intractable malignancy with high mortality. This is due in part to a high resistance to chemotherapeutics and radiation treatment conferred by diverse regulatory mechanisms. Among these, constituents of the nuclear envelope play a significant role in regulating oncogenesis and pancreatic tumor biology, and this review focuses on three specific components and their roles in cancer. The LINC complex is a nuclear envelope component formed by proteins with SUN and KASH domains that interact in the periplasmic space of the nuclear envelope. These interactions functionally and structurally couple the cytoskeleton to chromatin and facilitates gene regulation informed by cytoplasmic activity. Furthermore, cancer cell invasiveness is impacted by LINC complex biology. The nuclear lamina is adjacent to the inner nuclear membrane of the nuclear envelope and can actively regulate chromatin in addition to providing structural integrity to the nucleus. A disrupted lamina can impart biophysical compromise to nuclear structure and function, as well as form dysfunctional micronuclei that may lead to genomic instability and chromothripsis. In close relationship to the nuclear lamina is the nuclear pore complex, a large megadalton structure that spans both outer and inner membranes of the nuclear envelope. The nuclear pore complex mediates bidirectional nucleocytoplasmic transport and is comprised of specialized proteins called nucleoporins that are overexpressed in many cancers and are diagnostic markers for oncogenesis. Furthermore, recent demonstration of gene regulatory functions for discrete nucleoporins independent of their nuclear trafficking function suggests that these proteins may contribute more to malignant phenotypes beyond serving as biomarkers. The nuclear envelope is thus a complex, intricate regulator of cell signaling, with roles in pancreatic tumorigenesis and general oncogenic transformation.
Collapse
Affiliation(s)
| | - Randolph S. Faustino
- Genetics and Genomics, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
19
|
Lee H, Lee HJ, Jung JH, Shin EA, Kim SH. Melatonin disturbs SUMOylation-mediated crosstalk between c-Myc and nestin via MT1 activation and promotes the sensitivity of paclitaxel in brain cancer stem cells. J Pineal Res 2018; 65:e12496. [PMID: 29654697 DOI: 10.1111/jpi.12496] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
Here the underlying antitumor mechanism of melatonin and its potency as a sensitizer of paclitaxel was investigated in X02 cancer stem cells. Melatonin suppressed sphere formation and induced G2/M arrest in X02 cells expressing nestin, CD133, CXCR4, and SOX-2 as biomarkers of stemness. Furthermore, melatonin reduced the expression of CDK2, CDK4, cyclin D1, cyclin E, and c-Myc and upregulated cyclin B1 in X02 cells. Notably, genes of c-Myc related mRNAs were differentially expressed in melatonin-treated X02 cells by microarray analysis. Consistently, melatonin reduced the expression of c-Myc at mRNA and protein levels, which was blocked by MG132. Of note, overexpression of c-Myc increased the expression of nestin, while overexpression of nestin enhanced c-Myc through crosstalk despite different locations, nucleus, and cytoplasm. Interestingly, melatonin attenuated small ubiquitin-related modifier-1 (SUMO-1) more than SUMO-2 or SUMO-3 and disturbed nuclear translocation of nestin for direct binding to c-Myc by SUMOylation of SUMO-1 protein by immunofluorescence and immunoprecipitation. Also, melatonin reduced trimethylated histone H3K4me3 and H3K36me3 more than dimethylation in X02 cells by Western blotting and chromatin immunoprecipitation assay. Notably, melatonin upregulated MT1, not MT2, in X02 cells and melatonin receptor inhibitor luzindole blocked the ability of melatonin to decrease the expression of nestin, p-c-Myc(S62), and c-Myc. Furthermore, melatonin promoted cytotoxicity, sub-G1 accumulation, and apoptotic body formation by Paclitaxcel in X02 cells. Taken together, these findings suggest that melatonin inhibits stemness via suppression of c-Myc, nestin, and histone methylation via MT1 activation and promotes anticancer effect of Paclitaxcel in brain cancer stem cells.
Collapse
Affiliation(s)
- Hyemin Lee
- College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Hyo-Jung Lee
- College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Eun Ah Shin
- College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
20
|
Anyetei-Anum CS, Roggero VR, Allison LA. Thyroid hormone receptor localization in target tissues. J Endocrinol 2018; 237:R19-R34. [PMID: 29440347 PMCID: PMC5843491 DOI: 10.1530/joe-17-0708] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 12/28/2022]
Abstract
The thyroid hormone receptors, TRα1, TRβ1 and other subtypes, are members of the nuclear receptor superfamily that mediate the action of thyroid hormone signaling in numerous tissues to regulate important physiological and developmental processes. Their most well-characterized role is as ligand-dependent transcription factors; TRs bind thyroid hormone response elements in the presence or absence of thyroid hormone to facilitate the expression of target genes. Although primarily residing in the nucleus, TRα1 and TRβ1 shuttle rapidly between the nucleus and cytoplasm. We have identified multiple nuclear localization signals and nuclear export signals within TRα1 and TRβ1 that interact with importins and exportins, respectively, to mediate translocation across the nuclear envelope. More recently, enigmatic cytoplasmic functions have been ascribed to other TR subtypes, expanding the diversity of the cellular response to thyroid hormone. By integrating data on localization signal motifs, this review provides an overview of the complex interplay between TR's dynamic transport pathways and thyroid hormone signaling activities. We examine the variation in TR subtype response to thyroid hormone signaling, and what is currently known about regulation of the variety of tissue-specific localization patterns, including targeting to the nucleus, the mitochondria and the inner surface of the plasma membrane.
Collapse
Affiliation(s)
| | - Vincent R Roggero
- Department of BiologyCollege of William and Mary, Williamsburg, Virginia, USA
| | - Lizabeth A Allison
- Department of BiologyCollege of William and Mary, Williamsburg, Virginia, USA
| |
Collapse
|
21
|
Pandey D, Nomura Y, Rossberg MC, Hori D, Bhatta A, Keceli G, Leucker T, Santhanam L, Shimoda LA, Berkowitz D, Romer L. Hypoxia Triggers SENP1 (Sentrin-Specific Protease 1) Modulation of KLF15 (Kruppel-Like Factor 15) and Transcriptional Regulation of Arg2 (Arginase 2) in Pulmonary Endothelium. Arterioscler Thromb Vasc Biol 2018; 38:913-926. [PMID: 29472234 DOI: 10.1161/atvbaha.117.310660] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/06/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE KLF15 (Kruppel-like factor 15) has recently been shown to suppress activation of proinflammatory processes that contribute to atherogenesis in vascular smooth muscle, however, the role of KLF15 in vascular endothelial function is unknown. Arginase mediates inflammatory vasculopathy and vascular injury in pulmonary hypertension. Here, we tested the hypothesis that KLF15 is a critical regulator of hypoxia-induced Arg2 (arginase 2) transcription in human pulmonary microvascular endothelial cells (HPMEC). APPROACH AND RESULTS Quiescent HPMEC express ample amounts of full-length KLF15. HPMECs exposed to 24 hours of hypoxia exhibited a marked decrease in KLF15 protein levels and a reciprocal increase in Arg2 protein and mRNA. Chromatin immunoprecipitation indicated direct binding of KLF15 to the Arg2 promoter, which was relieved with HPMEC exposure to hypoxia. Furthermore, overexpression of KLF15 in HPMEC reversed hypoxia-induced augmentation of Arg2 abundance and arginase activity and rescued nitric oxide (NO) production. Ectopic KLF15 also reversed hypoxia-induced endothelium-mediated vasodilatation in isolated rat pulmonary artery rings. Mechanisms by which hypoxia regulates KLF15 abundance, stability, and compartmentalization to the nucleus in HPMEC were then investigated. Hypoxia triggered deSUMOylation of KLF15 by SENP1 (sentrin-specific protease 1), and translocation of KLF15 from nucleus to cytoplasm. CONCLUSIONS KLF15 is a critical regulator of pulmonary endothelial homeostasis via repression of endothelial Arg2 expression. KLF15 abundance and nuclear compartmentalization are regulated by SUMOylation/deSUMOylation-a hypoxia-sensitive process that is controlled by SENP1. Strategies including overexpression of KLF15 or inhibition of SENP1 may represent novel therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Deepesh Pandey
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD.
| | - Yohei Nomura
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Max C Rossberg
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Daijiro Hori
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Anil Bhatta
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Gizem Keceli
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Thorsten Leucker
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Lakshmi Santhanam
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Larissa A Shimoda
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Dan Berkowitz
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| | - Lewis Romer
- From the Departments of Anesthesiology and Critical Care Medicine (D.P., Y.N., M.C.R., D.H., A.B., L.S., D.B., L.R.), Cell Biology (L.R.), Biomedical Engineering (D.B., L.R.), and Pediatrics, and the Center for Cell Dynamics (L.R.), Division of Cardiology (G.K., T.L.), and Division of Pulmonary and Critical Care Medicine, Department of Medicine (L.A.S.), Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
22
|
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
|
23
|
Duheron V, Nilles N, Pecenko S, Martinelli V, Fahrenkrog B. Localisation of Nup153 and SENP1 to nuclear pore complexes is required for 53BP1-mediated DNA double-strand break repair. J Cell Sci 2017; 130:2306-2316. [PMID: 28576968 DOI: 10.1242/jcs.198390] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 05/28/2017] [Indexed: 12/28/2022] Open
Abstract
The nuclear basket of nuclear pore complexes (NPCs) is composed of three nucleoporins: Nup153, Nup50 and Tpr. Nup153 has a role in DNA double-strand break (DSB) repair by promoting nuclear import of 53BP1 (also known as TP53BP1), a mediator of the DNA damage response. Here, we provide evidence that loss of Nup153 compromises 53BP1 sumoylation, a prerequisite for efficient accumulation of 53BP1 at DSBs. Depletion of Nup153 resulted in reduced SUMO1 modification of 53BP1 and the displacement of the SUMO protease SENP1 from NPCs. Artificial tethering of SENP1 to NPCs restored non-homologous end joining (NHEJ) in the absence of Nup153 and re-established 53BP1 sumoylation. Furthermore, Nup50 and Tpr, the two other nuclear basket nucleoporins, also contribute to proper DSB repair, in a manner distinct from Nup153. Similar to the role of Nup153, Tpr is implicated in NHEJ and homologous recombination (HR), whereas loss of Nup50 only affects NHEJ. Despite the requirement of all three nucleoporins for accurate NHEJ, only Nup153 is needed for proper nuclear import of 53BP1 and SENP1-dependent sumoylation of 53BP1. Our data support the role of Nup153 as an important regulator of 53BP1 activity and efficient NHEJ.
Collapse
Affiliation(s)
- Vincent Duheron
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi 6041, Belgium
| | - Nadine Nilles
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi 6041, Belgium
| | - Sylvia Pecenko
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi 6041, Belgium
| | - Valérie Martinelli
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi 6041, Belgium
| | - Birthe Fahrenkrog
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi 6041, Belgium
| |
Collapse
|
24
|
A gene delivery system containing nuclear localization signal: Increased nucleus import and transfection efficiency with the assistance of RanGAP1. Acta Biomater 2017; 48:215-226. [PMID: 27816620 DOI: 10.1016/j.actbio.2016.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/02/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022]
Abstract
In the present report, a degradable gene delivery system (PAMS/DNA/10NLS) containing nucleus location signal peptide (NLS) was prepared. The agarose gel electrophoresis, particle size and zeta potential of PAMS/DNA/10NLS were similar to those of PAMS/DNA, which proved that NLS did not affect the interaction between PAMS and DNA. PAMS/DNA/10NLS exhibited marked extracellular and intracellular degradation under acidic conditions. The degradation was believed to allow NLS to come into contact with importins easily, which was able to mediate the nucleus import. With the help of NLS, PAMS/DNA/10NLS exhibited a higher transfection capability than PAMS/DNA. Moreover, the transfection of PAMS/DNA/10NLS was less dependent on the breakdown of the nucleus envelope than PAMS/DNA. Considering that GTPase-activating protein 1 (RanGAP1) was able to activate the endogenous GTPase, which was necessary for NLS-mediated nucleus import, RanGAP1 overexpressed cells (RanGAP1 cells) were produced. This result showed that RanGAP1 cells had higher GTPase activities than normal cells. Both the nucleus import and transfection efficiency of PAMS/DNA/10NLS were markedly higher in RanGAP1 cells than that in normal cells. The in vivo transfection results also showed that the transfection efficiency of PAMS/DNA/10NLS was higher in RanGAP1 pre-treated mice than that in normal mice. These findings showed that PAMS/DNA/10NLS is a promising gene delivery system with the assistance of RanGAP1. STATEMENT OF SIGNIFICANCE The present report describes the increased transfection efficiency of a degradable gene delivery system (PAMS/DNA/10NLS) containing nuclear location signal (NLS) with the assistance of GTPase-activating protein 1 (RanGAP1). The physicochemical properties of PAMS/DNA/10NLS were similar to those of PAMS/DNA. PAMS/DNA/10NLS exhibited great extracellular and intracellular degradations, which might allow NLS to contact with importins easily. With the help of NLS, PAMS/DNA/10NLS exhibited a higher transfection capability than PAMS/DNA. The transfection of PAMS/DNA/10NLS had less dependence on the breakdown of nuclear envelope. Both the nuclear import and transfection efficiency of PAMS/DNA/10NLS were higher in RanGAP1 overexpressed cells than that in normal cells. Moreover, the transfection efficiency of PAMS/DNA/10NLS was higher in RanGAP1 pre-treated mice than that in normal mice.
Collapse
|
25
|
Gan J, Qiao N, Strahan R, Zhu C, Liu L, Verma SC, Wei F, Cai Q. Manipulation of ubiquitin/SUMO pathways in human herpesviruses infection. Rev Med Virol 2016; 26:435-445. [DOI: 10.1002/rmv.1900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/03/2016] [Accepted: 07/25/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Jin Gan
- MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College; Fudan University; Shanghai China
| | - Niu Qiao
- Department of Medical Systems Biology, School of Basic Medical Sciences; Department of Translational Medicine, Shanghai Public Health Clinical Center; Institutes of Biomedical Sciences, Fudan University; Shanghai China
| | - Roxanne Strahan
- Department of Microbiology & Immunology; University of Nevada, Reno School of Medicine; Reno NV USA
| | - Caixia Zhu
- MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College; Fudan University; Shanghai China
| | - Lei Liu
- Department of Medical Systems Biology, School of Basic Medical Sciences; Department of Translational Medicine, Shanghai Public Health Clinical Center; Institutes of Biomedical Sciences, Fudan University; Shanghai China
| | - Subhash C. Verma
- Department of Microbiology & Immunology; University of Nevada, Reno School of Medicine; Reno NV USA
| | - Fang Wei
- ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology; Shanghai Jiao Tong University; Shanghai China
| | - Qiliang Cai
- MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College; Fudan University; Shanghai China
| |
Collapse
|
26
|
Audsley MD, Jans DA, Moseley GW. Roles of nuclear trafficking in infection by cytoplasmic negative-strand RNA viruses: paramyxoviruses and beyond. J Gen Virol 2016; 97:2463-2481. [PMID: 27498841 DOI: 10.1099/jgv.0.000575] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genome replication and virion production by most negative-sense RNA viruses (NSVs) occurs exclusively in the cytoplasm, but many NSV-expressed proteins undergo active nucleocytoplasmic trafficking via signals that exploit cellular nuclear transport pathways. Nuclear trafficking has been reported both for NSV accessory proteins (including isoforms of the rabies virus phosphoprotein, and V, W and C proteins of paramyxoviruses) and for structural proteins. Trafficking of the former is thought to enable accessory functions in viral modulation of antiviral responses including the type I IFN system, but the intranuclear roles of structural proteins such as nucleocapsid and matrix proteins, which have critical roles in extranuclear replication and viral assembly, are less clear. Nevertheless, nuclear trafficking of matrix protein has been reported to be critical for efficient production of Nipah virus and Respiratory syncytial virus, and nuclear localization of nucleocapsid protein of several morbilliviruses has been linked to mechanisms of immune evasion. Together, these data point to the nucleus as a significant host interface for viral proteins during infection by NSVs with otherwise cytoplasmic life cycles. Importantly, several lines of evidence now suggest that nuclear trafficking of these proteins may be critical to pathogenesis and thus could provide new targets for vaccine development and antiviral therapies.
Collapse
Affiliation(s)
- Michelle D Audsley
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Gregory W Moseley
- Department of Biochemistry and Molecular Biology, BIO21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC 3000, Australia
| |
Collapse
|
27
|
Alvarez MJ, Shen Y, Giorgi FM, Lachmann A, Ding BB, Ye BH, Califano A. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat Genet 2016; 48:838-47. [PMID: 27322546 PMCID: PMC5040167 DOI: 10.1038/ng.3593] [Citation(s) in RCA: 580] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/23/2016] [Indexed: 01/05/2023]
Abstract
Identifying the multiple dysregulated oncoproteins that contribute to tumorigenesis in a given patient is crucial for developing personalized treatment plans. However, accurate inference of aberrant protein activity in biological samples is still challenging as genetic alterations are only partially predictive and direct measurements of protein activity are generally not feasible. To address this problem we introduce and experimentally validate a new algorithm, virtual inference of protein activity by enriched regulon analysis (VIPER), for accurate assessment of protein activity from gene expression data. We used VIPER to evaluate the functional relevance of genetic alterations in regulatory proteins across all samples in The Cancer Genome Atlas (TCGA). In addition to accurately infer aberrant protein activity induced by established mutations, we also identified a fraction of tumors with aberrant activity of druggable oncoproteins despite a lack of mutations, and vice versa. In vitro assays confirmed that VIPER-inferred protein activity outperformed mutational analysis in predicting sensitivity to targeted inhibitors.
Collapse
Affiliation(s)
- Mariano J. Alvarez
- Department of Systems Biology, Columbia University, New York, USA
- DarwinHealth Inc., New York, USA
| | - Yao Shen
- Department of Systems Biology, Columbia University, New York, USA
- DarwinHealth Inc., New York, USA
| | | | | | - B. Belinda Ding
- Department of Cell Biology, Albert Einstein College of Medicine, New York, USA
| | - B. Hilda Ye
- Department of Cell Biology, Albert Einstein College of Medicine, New York, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, USA
- Department of Biomedical Informatics, Columbia University, New York, USA
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, USA
- Institute for Cancer Genetics, Columbia University, New York, USA
- Motor Neuron Center, Columbia University, New York, USA
- Columbia Initiative in Stem Cells, Columbia University, New York, USA
| |
Collapse
|
28
|
Cahill MA, Jazayeri JA, Catalano SM, Toyokuni S, Kovacevic Z, Richardson DR. The emerging role of progesterone receptor membrane component 1 (PGRMC1) in cancer biology. Biochim Biophys Acta Rev Cancer 2016; 1866:339-349. [PMID: 27452206 DOI: 10.1016/j.bbcan.2016.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 07/17/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023]
Abstract
Progesterone receptor membrane component 1 (PGRMC1) is a multi-functional protein with a heme-binding moiety related to that of cytochrome b5, which is a putative progesterone receptor. The recently solved PGRMC1 structure revealed that heme-binding involves coordination by a tyrosinate ion at Y113, and induces dimerization which is stabilized by hydrophobic stacking of heme on adjacent monomers. Dimerization is required for association with cytochrome P450 (cyP450) enzymes, which mediates chemoresistance to doxorubicin and may be responsible for PGRMC1's anti-apoptotic activity. Here we review the multiple attested involvement of PGRMC1 in diverse functions, including regulation of cytochrome P450, steroidogenesis, vesicle trafficking, progesterone signaling and mitotic spindle and cell cycle regulation. Its wide range of biological functions is attested to particularly by its emerging association with cancer and progesterone-responsive female reproductive tissues. PGRMC1 exhibits all the hallmarks of a higher order nexus signal integration hub protein. It appears capable of acting as a detector that integrates information from kinase/phosphatase pathways with heme and CO levels and probably redox status.
Collapse
Affiliation(s)
- Michael A Cahill
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia.
| | - Jalal A Jazayeri
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Susan M Catalano
- Cognition Therapeutics Inc., Pittsburgh, PA 15203, United States
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
29
|
Safdar K, Gu A, Xu X, Au V, Taylor J, Flibotte S, Moerman DG, Maine EM. UBR-5, a Conserved HECT-Type E3 Ubiquitin Ligase, Negatively Regulates Notch-Type Signaling in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2016; 6:2125-34. [PMID: 27185398 PMCID: PMC4938665 DOI: 10.1534/g3.116.027805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022]
Abstract
Notch-type signaling mediates cell-cell interactions important for animal development. In humans, reduced or inappropriate Notch signaling activity is associated with various developmental defects and disease states, including cancers. Caenorhabditis elegans expresses two Notch-type receptors, GLP-1 and LIN-12. GLP-1 mediates several cell-signaling events in the embryo and promotes germline proliferation in the developing and adult gonad. LIN-12 acts redundantly with GLP-1 in certain inductive events in the embryo and mediates several cell-cell interactions during larval development. Recovery of genetic suppressors and enhancers of glp-1 or lin-12 loss- or gain-of-function mutations has identified numerous regulators of GLP-1 and LIN-12 signaling activity. Here, we report the molecular identification of sog-1, a gene identified in screens for recessive suppressors of conditional glp-1 loss-of-function mutations. The sog-1 gene encodes UBR-5, the sole C. elegans member of the UBR5/Hyd family of HECT-type E3 ubiquitin ligases. Molecular and genetic analyses indicate that the loss of ubr-5 function suppresses defects caused by reduced signaling via GLP-1 or LIN-12. In contrast, ubr-5 mutations do not suppress embryonic or larval lethality associated with mutations in a downstream transcription factor, LAG-1. In the gonad, ubr-5 acts in the receiving cells (germ cells) to limit GLP-1 signaling activity. SEL-10 is the F-box component of SCF(SEL-10) E3 ubiquitin-ligase complex that promotes turnover of Notch intracellular domain. UBR-5 acts redundantly with SEL-10 to limit Notch signaling in certain tissues. We hypothesize that UBR-5 activity limits Notch-type signaling by promoting turnover of receptor or limiting its interaction with pathway components.
Collapse
Affiliation(s)
- Komal Safdar
- Department of Biology, Syracuse University, New York 13244
| | - Anniya Gu
- Department of Biology, Syracuse University, New York 13244
| | - Xia Xu
- Department of Biology, Syracuse University, New York 13244
| | - Vinci Au
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Jon Taylor
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Stephane Flibotte
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Donald G Moerman
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | | |
Collapse
|
30
|
Halvorsen AR, Silwal-Pandit L, Meza-Zepeda LA, Vodak D, Vu P, Sagerup C, Hovig E, Myklebost O, Børresen-Dale AL, Brustugun OT, Helland Å. TP53 Mutation Spectrum in Smokers and Never Smoking Lung Cancer Patients. Front Genet 2016; 7:85. [PMID: 27242894 PMCID: PMC4863128 DOI: 10.3389/fgene.2016.00085] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/25/2016] [Indexed: 01/27/2023] Open
Abstract
Background:TP53 mutations are among the most common mutations found in lung cancers, identified as an independent prognostic factor in many types of cancers. The purpose of this study was to investigate the frequency and prognostic impact of TP53 mutations in never-smokers and in different histological subtypes of lung cancer. Methods: We analyzed tumor tissue from 394 non-small cell carcinomas including adenocarcinomas (n = 229), squamous cell carcinomas (n = 112), large cell carcinomas (n = 30), and others (n = 23) for mutations in TP53 by the use of Sanger sequencing (n = 394) and next generation sequencing (n = 100). Results:TP53 mutations were identified in 47.2% of the samples, with the highest frequency (65%) of mutations among squamous cell carcinomas. Among never-smokers, 36% carried a TP53 mutation, identified as a significant independent negative prognostic factor in this subgroup. For large cell carcinomas, a significantly prolonged progression free survival was found for those carrying a TP53 mutation. In addition, the frequency of frameshift mutations was doubled in squamous cell carcinomas (20.3%) compared to adenocarcinomas (9.1%). Conclusion:TP53 mutation patterns differ between the histological subgroups of lung cancers, and are also influenced by smoking history. This indicates that the histological subtypes in lung cancer are genetically different, and that smoking-induced TP53 mutations may have a different biological impact than TP53 mutations occurring in never-smokers.
Collapse
Affiliation(s)
- Ann R Halvorsen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital Oslo, Norway
| | - Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital Oslo, Norway
| | - Leonardo A Meza-Zepeda
- Department of Tumour Biology, Institute for Cancer Research, Oslo University HospitalOslo, Norway; Genomics Core Facility, Department of Core Facilities, Institute for Cancer Research, Oslo University HospitalOslo, Norway
| | - Daniel Vodak
- Department of Tumour Biology, Institute for Cancer Research, Oslo University Hospital Oslo, Norway
| | - Phuong Vu
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital Oslo, Norway
| | - Camilla Sagerup
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital Oslo, Norway
| | - Eivind Hovig
- Department of Tumour Biology, Institute for Cancer Research, Oslo University HospitalOslo, Norway; Department of Informatics, University of OsloOslo, Norway; Institute of Cancer Genetics and Informatics, Oslo University HospitalOslo, Norway
| | - Ola Myklebost
- Department of Tumour Biology, Institute for Cancer Research, Oslo University Hospital Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium HospitalOslo, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of OsloOslo, Norway
| | - Odd T Brustugun
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium HospitalOslo, Norway; Department of Oncology, Oslo University Hospital - The Norwegian Radium HospitalOslo, Norway
| | - Åslaug Helland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium HospitalOslo, Norway; Department of Oncology, Oslo University Hospital - The Norwegian Radium HospitalOslo, Norway
| |
Collapse
|
31
|
Masoumi KC, Marfany G, Wu Y, Massoumi R. Putative role of SUMOylation in controlling the activity of deubiquitinating enzymes in cancer. Future Oncol 2016; 12:565-74. [PMID: 26777062 DOI: 10.2217/fon.15.320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Deubiquitinating enzymes (DUBs) are specialized proteins that can recognize ubiquitinated proteins, and after direct interaction, deconjugate monomeric or polymeric ubiquitin chains, thus changing the fate of the substrates. This process is instrumental in mediating or changing downstream signaling pathways. Beside mutations and alterations in their expression levels, the activity and stability of deubiquitinating enzymes is vital for their function. SUMOylations consist of the conjugation of the small peptide SUMO to protein substrates which is very similar to ubiquitination in the mechanistic and machinery required. In this review, we will focus on how SUMOylation can regulate DUB enzymatic activity, stability or DUB interaction with partners and substrates, in cancer. Furthermore, we will discuss the impact of these recent findings in the identification of new potential tools for efficient anticancer treatment strategies.
Collapse
Affiliation(s)
- Katarzyna C Masoumi
- Department of Laboratory Medicine, Medicon Village, Lund University, 22381 Lund, Sweden
| | - Gemma Marfany
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain.,Institut de Biomedicina (IBUB), Universitat de Barcelona, 08007 Barcelona, Spain.,CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Yingli Wu
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ramin Massoumi
- Department of Laboratory Medicine, Medicon Village, Lund University, 22381 Lund, Sweden
| |
Collapse
|
32
|
Harrison JS, Jacobs TM, Houlihan K, Van Doorslaer K, Kuhlman B. UbSRD: The Ubiquitin Structural Relational Database. J Mol Biol 2015; 428:679-687. [PMID: 26392143 DOI: 10.1016/j.jmb.2015.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/08/2015] [Accepted: 09/13/2015] [Indexed: 10/23/2022]
Abstract
The structurally defined ubiquitin-like homology fold (UBL) can engage in several unique protein-protein interactions and many of these complexes have been characterized with high-resolution techniques. Using Rosetta's structural classification tools, we have created the Ubiquitin Structural Relational Database (UbSRD), an SQL database of features for all 509 UBL-containing structures in the PDB, allowing users to browse these structures by protein-protein interaction and providing a platform for quantitative analysis of structural features. We used UbSRD to define the recognition features of ubiquitin (UBQ) and SUMO observed in the PDB and the orientation of the UBQ tail while interacting with certain types of proteins. While some of the interaction surfaces on UBQ and SUMO overlap, each molecule has distinct features that aid in molecular discrimination. Additionally, we find that the UBQ tail is malleable and can adopt a variety of conformations upon binding. UbSRD is accessible as an online resource at rosettadesign.med.unc.edu/ubsrd.
Collapse
Affiliation(s)
- Joseph S Harrison
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tim M Jacobs
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin Houlihan
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Koenraad Van Doorslaer
- DNA Tumor Virus Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
33
|
Calistri A, Munegato D, Toffoletto M, Celestino M, Franchin E, Comin A, Sartori E, Salata C, Parolin C, Palù G. Functional Interaction Between the ESCRT-I Component TSG101 and the HSV-1 Tegument Ubiquitin Specific Protease. J Cell Physiol 2015; 230:1794-806. [PMID: 25510868 DOI: 10.1002/jcp.24890] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 12/11/2014] [Indexed: 02/01/2023]
Abstract
Similar to phosphorylation, transient conjugation of ubiquitin to target proteins (ubiquitination) mediated by the concerted action of ubiquitin ligases and de-ubiquitinating enzymes (DUBs) can affect substrate function. As obligate intracellular parasites, viruses rely on different cellular pathways for their own replication and the well conserved ubiquitin conjugating/de-conjugating system is not an exception. Viruses not only usurp the host proteins involved in the ubiquitination/de-ubiquitination process, but they also encode their own ubiquitin ligases and DUBs. Here we report that an N-terminal variant of the herpes simplex virus (HSV) type-1 large tegument protein VP1/2 (VP1/2(1-767)), encompassing an active DUB domain (herpesvirus tegument ubiquitin specific protease, htUSP), and TSG101, a component of the endosomal sorting complex required for transport (ESCRT)-I, functionally interact. In particular, VP1/2(1-767) modulates TSG101 ubiquitination and influences its intracellular distribution. Given the role played by the ESCRT machinery in crucial steps of both cellular pathways and viral life cycle, the identification of TSG101 as a cellular target for the HSV-1 specific de-ubiquitinating enzyme contributes to the clarification of the still under debate function of viral encoded DUBs highly conserved throughout the Herpesviridae family.
Collapse
Affiliation(s)
- A Calistri
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gan J, Wang C, Jin Y, Guo Y, Xu F, Zhu Q, Ding L, Shang H, Wang J, Wei F, Cai Q, Robertson ES. Proteomic profiling identifies the SIM-associated complex of KSHV-encoded LANA. Proteomics 2015; 15:2023-37. [PMID: 25894481 PMCID: PMC5868752 DOI: 10.1002/pmic.201400624] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 02/11/2015] [Accepted: 04/17/2015] [Indexed: 11/08/2022]
Abstract
The Kaposi's sarcoma-associated herpesvirus (KSHV) encoded latent nuclear antigen latency-associated nuclear antigen (LANA) plays an essential role in viral episome maintenance. LANA also contributes to DNA replication and tumorigenesis during latency. Recent studies suggested that LANA was involved in regulation of SUMOylation, which results in chromatin silencing. To examine the pleiotropic effects of LANA protein on host cell gene expression, we utilized MS analysis to identify cellular proteins associated with the small ubiquitin related modifier (SUMO) interacting motif of LANA (LANA(SIM)). In addition to the six bands identified as substantially associated with LANA(SIM), 151 proteins were positively identified by MS/MS analysis. Compared with previous proteomic analysis of the N- and C-truncated mutants of LANA (LANA(NC)), our results revealed that a complex of specific proteins with relatively high SUMOylation and SIM motifs is associated with LANA(SIM). Intriguingly, consistent with our previous report that identified KAP1 as a key component, the in vitro SUMO-2-modified isoform has a substantially higher affinity with LANA(SIM) than the SUMO-1-modified isoform. Moreover, via cluster and pathway analysis, we proposed a hypothetical model for the LANA(SIM) regulatory circuit involving aberrant SUMOylation of cell cycle (particular mitotic), DNA unwinding and replication, and pre-mRNA/mRNA processing related proteins. This study provides a SUMOylated and non-SUMOylated proteome profile of LANA(SIM) -associated complex and facilitates our understanding that viral-mediated gene regulation through SUMOylation is important for Kaposi's sarcoma-associated herpesvirus persistence and pathogenesis.
Collapse
Affiliation(s)
- Jin Gan
- MOE& MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Chong Wang
- MOE& MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Yanling Jin
- ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Yi Guo
- Department of Gynecology, Key laboratory of AIDS immunology of Ministry of Health, First Affiliated Hospital of China Medical University, Shenyang 110000, P. R. China
| | - Feng Xu
- Department of Biochemistry and Center for Genome Science, The University of Hong Kong, Hong Kong, P. R. China
| | - Qing Zhu
- MOE& MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Ling Ding
- MOE& MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Hong Shang
- Department of Gynecology, Key laboratory of AIDS immunology of Ministry of Health, First Affiliated Hospital of China Medical University, Shenyang 110000, P. R. China
| | - Junwen Wang
- Department of Biochemistry and Center for Genome Science, The University of Hong Kong, Hong Kong, P. R. China
| | - Fang Wei
- ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Qiliang Cai
- MOE& MOH Key Laboratory of Medical Molecular Virology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Erle S Robertson
- Department of Microbiology and Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia 19104, USA
| |
Collapse
|
35
|
Lamoliatte F, Caron D, Durette C, Mahrouche L, Maroui MA, Caron-Lizotte O, Bonneil E, Chelbi-Alix MK, Thibault P. Large-scale analysis of lysine SUMOylation by SUMO remnant immunoaffinity profiling. Nat Commun 2014; 5:5409. [PMID: 25391492 DOI: 10.1038/ncomms6409] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 09/29/2014] [Indexed: 01/11/2023] Open
Abstract
Small ubiquitin-related modifiers (SUMO) are evolutionarily conserved ubiquitin-like proteins that regulate several cellular processes including cell cycle progression, intracellular trafficking, protein degradation and apoptosis. Despite the importance of protein SUMOylation in different biological pathways, the global identification of acceptor sites in complex cell extracts remains a challenge. Here we generate a monoclonal antibody that enriches for peptides containing SUMO remnant chains following tryptic digestion. We identify 954 SUMO3-modified lysine residues on 538 proteins and profile by quantitative proteomics the dynamic changes of protein SUMOylation following proteasome inhibition. More than 86% of these SUMOylation sites have not been reported previously, including 5 sites on the tumour suppressor parafibromin (CDC73). The modification of CDC73 at K136 affects its nuclear retention within PML nuclear bodies on proteasome inhibition. In contrast, a CDC73 K136R mutant translocates to the cytoplasm under the same conditions, further demonstrating the effectiveness of our method to characterize the dynamics of lysine SUMOylation.
Collapse
Affiliation(s)
- Frédéric Lamoliatte
- 1] Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7 [2] Department of Chemistry, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | - Danielle Caron
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | - Chantal Durette
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | - Louiza Mahrouche
- 1] Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7 [2] Department of Biochemistry, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | | | - Olivier Caron-Lizotte
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | - Eric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| | | | - Pierre Thibault
- 1] Institute for Research in Immunology and Cancer, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7 [2] Department of Chemistry, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7 [3] Department of Biochemistry, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, Québec, Canada H3C 3J7
| |
Collapse
|
36
|
Ehrlichia chaffeensis exploits host SUMOylation pathways to mediate effector-host interactions and promote intracellular survival. Infect Immun 2014; 82:4154-68. [PMID: 25047847 DOI: 10.1128/iai.01984-14] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Ehrlichia chaffeensis is an obligately intracellular Gram-negative bacterium that selectively infects mononuclear phagocytes. We recently reported that E. chaffeensis utilizes a type 1 secretion (T1S) system to export tandem repeat protein (TRP) effectors and demonstrated that these effectors interact with a functionally diverse array of host proteins. By way of these interactions, TRP effectors modulate host cell functions; however, the molecular basis of these interactions and their roles in ehrlichial pathobiology are not well defined. In this study, we describe the first bacterial protein posttranslational modification (PTM) by the small ubiquitin-like modifier (SUMO). The E. chaffeensis T1S effector TRP120 is conjugated to SUMO at a carboxy-terminal canonical consensus SUMO conjugation motif in vitro and in human cells. In human cells, TRP120 was selectively conjugated with SUMO2/3 isoforms. Disruption of TRP120 SUMOylation perturbed interactions with known host proteins, through predicted SUMO interaction motif-dependent and -independent mechanisms. E. chaffeensis infection did not result in dramatic changes in the global host SUMOylated protein profile, but a robust colocalization of predominately SUMO1 with ehrlichial inclusions was observed. Inhibiting the SUMO pathway with a small-molecule inhibitor had a significant impact on E. chaffeensis replication and recruitment of the TRP120-interacting protein polycomb group ring finger protein 5 (PCGF5) to the inclusion, indicating that the SUMO pathway is critical for intracellular survival. This study reveals the novel exploitation of the SUMO pathway by Ehrlichia, which facilitates effector-eukaryote interactions necessary to usurp the host and create a permissive intracellular niche.
Collapse
|