1
|
Wang J, Topilin I, Feofilova A, Shao M, Wang Y. Cooperative Intelligent Transport Systems: The Impact of C-V2X Communication Technologies on Road Safety and Traffic Efficiency. SENSORS (BASEL, SWITZERLAND) 2025; 25:2132. [PMID: 40218643 PMCID: PMC11990983 DOI: 10.3390/s25072132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
The advancement of intelligent road transport represents a promising direction in the evolution of transportation systems, aimed at improving road safety and reducing traffic accidents. The integration of artificial intelligence, sensors, and machine vision systems enables autonomous vehicles (AVs) to rapidly adapt to changes in the road environment, minimizing human error and significantly reducing collision risks. These technologies provide continuous and highly precise control, including adaptive acceleration, braking, and maneuvering, thereby enhancing overall road safety. Connected vehicles utilizing C-V2X (Cellular Vehicle-to-Everything) communication primarily feature real-time operation, safety, and stability. However, communication flaws, such as signal fading, time delays, packet loss, and malicious network attacks, can affect vehicle-to-vehicle interactions in cooperative intelligent transport systems (C-ITSs). This study explores how C-V2X technology, compared to traditional DSRC, improves communication latency and enhances vehicle communication efficiency. Using SUMO simulations, various traffic scenarios were modeled with different autonomous vehicle penetration rates and communication technologies, focusing on traffic conflict rates, travel time, and communication performance. The results demonstrated that C-V2X reduced latency by over 99% compared to DSRC, facilitating faster communication between vehicles and contributing to a 38% reduction in traffic conflicts at 60% AV penetration. Traffic flow and safety improved with increased AV penetration, particularly in congested conditions. While C-V2X offers substantial benefits, challenges such as data packet loss, communication delays, and security vulnerabilities must be addressed to fully realize its potential. Future advancements in 5G and subsequent wireless communication technologies are expected to further reduce latency and enhance the effectiveness of C-ITSs. This study underscores the potential of C-V2X to enhance collision avoidance, alleviate congestion, and improve traffic management, while also contributing to the development of more reliable and efficient transportation systems. The continued refinement of simulation models and collaboration among stakeholders will be crucial to addressing the challenges in CAV integration and realizing the full benefits of connected transportation systems in smart cities.
Collapse
Affiliation(s)
- Jingwen Wang
- Don School, International Education College, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (Y.W.)
| | - Ivan Topilin
- Faculty of Road and Transportation, Don State Technical University, 1 Gagarin sq., Rostov-on-Don 344000, Russia; (I.T.); (A.F.)
| | - Anastasia Feofilova
- Faculty of Road and Transportation, Don State Technical University, 1 Gagarin sq., Rostov-on-Don 344000, Russia; (I.T.); (A.F.)
| | - Mengru Shao
- Urban and Data Science, Graduate School of Advanced Science and Engineering, Hiroshima University, Higashi-Hiroshima 739-8511, Japan
| | - Yadong Wang
- Don School, International Education College, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (Y.W.)
| |
Collapse
|
2
|
Baytshtok V, DiMattia MA, Lima CD. Structural basis for a nucleoporin exportin complex between RanBP2, SUMO1-RanGAP1, the E2 Ubc9, Crm1 and the Ran GTPase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616749. [PMID: 39763778 PMCID: PMC11703149 DOI: 10.1101/2024.10.04.616749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
The human nucleoporin RanBP2/Nup358 interacts with SUMO1-modified RanGAP1 and the SUMO E2 Ubc9 at the nuclear pore complex (NPC) to promote export and disassembly of exportin Crm1/Ran(GTP)/cargo complexes. In mitosis, RanBP2/SUMO1-RanGAP1/Ubc9 remains intact after NPC disassembly and is recruited to kinetochores and mitotic spindles by Crm1 where it contributes to mitotic progression. Interestingly, RanBP2 binds SUMO1-RanGAP1/Ubc9 via motifs that also catalyze SUMO E3 ligase activity. Here, we resolve cryo-EM structures of a RanBP2 C-terminal fragment in complex with Crm1, SUMO1-RanGAP1/Ubc9, and two molecules of Ran(GTP). These structures reveal several unanticipated interactions with Crm1 including a nuclear export signal (NES) for RanGAP1, the deletion of which mislocalizes RanGAP1 and the Ran GTPase in cells. Our structural and biochemical results support models in which RanBP2 E3 ligase activity is dependent on Crm1, the RanGAP1 NES and Ran GTPase cycling.
Collapse
Affiliation(s)
- Vladimir Baytshtok
- Structural Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065
- These authors contributed equally
| | - Michael A DiMattia
- Structural Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065
- Current address: Schrödinger New York, 1540 Broadway, 24th Floor, New York, NY 10036, USA
- These authors contributed equally
| | - Christopher D Lima
- Structural Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065
- Howard Hughes Medical Institute, 1275 York Avenue, New York, NY 10065
| |
Collapse
|
3
|
Fertig B, Ling J, Nollet EE, Dobi S, Busiau T, Ishikawa K, Yamada K, Lee A, Kho C, Wills L, Tibbo AJ, Scott M, Grant K, Campbell KS, Birks EJ, MacQuaide N, Hajjar R, Smith GL, van der Velden J, Baillie GS. SUMOylation does not affect cardiac troponin I stability but alters indirectly the development of force in response to Ca 2. FEBS J 2022; 289:6267-6285. [PMID: 35633070 PMCID: PMC9588612 DOI: 10.1111/febs.16537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 03/21/2022] [Accepted: 05/26/2022] [Indexed: 11/27/2022]
Abstract
Post-translational modification of the myofilament protein troponin I by phosphorylation is known to trigger functional changes that support enhanced contraction and relaxation of the heart. We report for the first time that human troponin I can also be modified by SUMOylation at lysine 177. Functionally, TnI SUMOylation is not a factor in the development of passive and maximal force generation in response to calcium, however this modification seems to act indirectly by preventing SUMOylation of other myofilament proteins to alter calcium sensitivity and cooperativity of myofilaments. Utilising a novel, custom SUMO site-specific antibody that recognises only the SUMOylated form of troponin I, we verify that this modification occurs in human heart and that it is upregulated during disease.
Collapse
Affiliation(s)
- Bracy Fertig
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| | - Jiayue Ling
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| | - Edgar E. Nollet
- Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular SciencesVrije Universiteit AmsterdamThe Netherlands
| | - Sara Dobi
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| | - Tara Busiau
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| | | | - Kelly Yamada
- Cardiovascular Research CentreIcahn School of MedicineNew YorkNYUSA
| | - Ahyoung Lee
- Cardiovascular Research CentreIcahn School of MedicineNew YorkNYUSA
| | - Changwon Kho
- Division of Applied MedicinePusan National UniversityKorea
| | - Lauren Wills
- Department of NeuroscienceIchan School of MedicineNew YorkNYUSA
| | - Amy J. Tibbo
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| | - Mark Scott
- INSERM, U1016, Institut CochinParisFrance
| | - Kirsten Grant
- Department of Clinical BiochemistryGlasgow Royal InfirmaryUK
| | - Kenneth S. Campbell
- Department of PhysiologyUniversity of KentuckyLexingtonKYUSA
- Division of Cardiovasuclar MedicineUniversity of KentuckyLexingtonKYUSA
| | - Emma J. Birks
- Division of Cardiovasuclar MedicineUniversity of KentuckyLexingtonKYUSA
| | - Niall MacQuaide
- School of Health and Life SciencesGlasgow Caledonian UniversityUK
| | | | - Godfrey L. Smith
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular SciencesVrije Universiteit AmsterdamThe Netherlands
- Netherlands Heart InstituteUtrechtThe Netherlands
| | - George S. Baillie
- Institute of Cardiovascular and Medical Sciences, College of Veterinary, Medical and Life SciencesGlasgow UniversityUK
| |
Collapse
|
4
|
Srivastava M, Sadanandom A, Srivastava AK. Towards understanding the multifaceted role of SUMOylation in plant growth and development. PHYSIOLOGIA PLANTARUM 2021; 171:77-85. [PMID: 32880960 DOI: 10.1111/ppl.13204] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 06/11/2023]
Abstract
Post-translational modifications (PTMs) play a critical role in regulating plant growth and development through the modulation of protein functionality and its interaction with its partners. Analysis of the functional implication of PTMs on plant cellular signalling presents grand challenges in understanding their significance. Proteins decorated or modified with another chemical group or polypeptide play a significant role in regulating physiological processes as compared with non-decorated or non-modified proteins. In the past decade, SUMOylation has been emerging as a potent PTM influencing the adaptability of plants to growth, in response to various environmental cues. Deciphering the SUMO-mediated regulation of plant stress responses and its consequences is required to understand the mechanism underneath. Here, we will discuss the recent advances in the role and significance of SUMOylation in plant growth, development and stress response.
Collapse
Affiliation(s)
| | - Ari Sadanandom
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| | | |
Collapse
|
5
|
Viral DNA Binding Protein SUMOylation Promotes PML Nuclear Body Localization Next to Viral Replication Centers. mBio 2020; 11:mBio.00049-20. [PMID: 32184235 PMCID: PMC7078464 DOI: 10.1128/mbio.00049-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human adenoviruses (HAdVs) have developed mechanisms to manipulate cellular antiviral measures to ensure proper DNA replication, with detailed processes far from being understood. Host cells repress incoming viral genomes through a network of transcriptional regulators that normally control cellular homeostasis. The nuclear domains involved are promyelocytic leukemia protein nuclear bodies (PML-NBs), interferon-inducible, dot-like nuclear structures and hot spots of SUMO posttranslational modification (PTM). In HAdV-infected cells, such SUMO factories are found in close proximity to newly established viral replication centers (RCs) marked by the adenoviral DNA binding protein (DBP) E2A. Here, we show that E2A is a novel target of host SUMOylation, leading to PTMs supporting E2A function in promoting productive infection. Our data show that SUMOylated E2A interacts with PML. Decreasing SUMO-E2A protein levels by generating HAdV variants mutated in the three main SUMO conjugation motifs (SCMs) led to lower numbers of viral RCs and PML-NBs, and these two structures were no longer next to each other. Our data further indicate that SUMOylated E2A binds the host transcription factor Sp100A, promoting HAdV gene expression, and represents the molecular bridge between PML tracks and adjacent viral RCs. Consequently, E2A SCM mutations repressed late viral gene expression and progeny production. These data highlight a novel mechanism used by the virus to benefit from host antiviral responses by exploiting the cellular SUMO conjugation machinery.IMPORTANCE PML nuclear bodies (PML-NBs) are implicated in general antiviral defense based on recruiting host restriction factors; however, it is not understood so far why viruses would establish viral replication centers (RCs) juxtaposed to such "antiviral" compartments. To understand this enigma, we investigate the cross talk between PML-NB components and viral RCs to find the missing link connecting both compartments to promote efficient viral replication and gene expression. Taken together, the current concept is more intricate than originally believed, since viruses apparently take advantage of several specific PML-NB-associated proteins to promote productive infection. Simultaneously, they efficiently inhibit antiviral measures to maintain the viral infectious program. Our data provide evidence that SUMOylation of the viral RC marker protein E2A represents the basis of this virus-host interface and regulates various downstream events to support HAdV productive infection. These results are the basis of our current attempts to generate and screen for specific E2A SUMOylation inhibitors to constitute novel therapeutic approaches to limit and prevent HAdV-mediated diseases and mortality of immunosuppressed patients.
Collapse
|
6
|
Yu HI, Hsu T, Maruyama EO, Paschen W, Yang W, Hsu W. The requirement of SUMO2/3 for SENP2 mediated extraembryonic and embryonic development. Dev Dyn 2020; 249:237-244. [PMID: 31625212 PMCID: PMC7027852 DOI: 10.1002/dvdy.125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/23/2022] Open
Abstract
Small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is essential for the development of healthy placenta. The loss of SENP2 causes severe placental deficiencies and leads to embryonic death that is associated with heart and brain deformities. However, tissue-specific disruption of SENP2 demonstrates its dispensable role in embryogenesis and the embryonic defects are secondary to placental insufficiency. SENP2 regulates SUMO1 modification of Mdm2, which controls p53 activities critical for trophoblast cell proliferation and differentiation. Here we use genetic analyses to examine the involvement of SUMO2 and SUMO3 for SENP2-mediated placentation. The results indicate that hyper-SUMOylation caused by SENP2 deficiency can be compensated by reducing the level of SUMO modifiers. The placental deficiencies caused by the loss of SENP2 can be alleviated by the inactivation of gene encoding SUMO2 or SUMO3. Our findings demonstrate that SENP2 genetically interacts with SUMO2 and SUMO3 pivotal for the development of three major trophoblast layers. The alleviation of placental defects in the SENP2 knockouts further leads to the proper formation of the heart structures, including atrioventricular cushion and myocardium. SUMO2 and SUMO3 modifications regulate placentation and organogenesis mediated by SENP2.
Collapse
Affiliation(s)
- H‐M Ivy Yu
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
| | - Trunee Hsu
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
- Pittsford Mendon High SchoolPittsfordNew York
| | - Eri O Maruyama
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
| | - Wulf Paschen
- Department of AnesthesiologyDuke UniversityDurhamNorth Carolina
| | - Wei Yang
- Department of AnesthesiologyDuke UniversityDurhamNorth Carolina
| | - Wei Hsu
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
- Department of Biomedical GeneticsUniversity of Rochester Medical CenterRochesterNew York
- Stem Cell and Regenerative Medicine Institute, University of Rochester Medical CenterRochesterNew York
| |
Collapse
|
7
|
Wesalo JS, Luo J, Morihiro K, Liu J, Deiters A. Phosphine-Activated Lysine Analogues for Fast Chemical Control of Protein Subcellular Localization and Protein SUMOylation. Chembiochem 2020; 21:141-148. [PMID: 31664790 PMCID: PMC6980333 DOI: 10.1002/cbic.201900464] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/03/2019] [Indexed: 11/06/2022]
Abstract
The Staudinger reduction and its variants have exceptional compatibility with live cells but can be limited by slow kinetics. Herein we report new small-molecule triggers that turn on proteins through a Staudinger reduction/self-immolation cascade with substantially improved kinetics and yields. We achieved this through site-specific incorporation of a new set of azidobenzyloxycarbonyl lysine derivatives in mammalian cells. This approach allowed us to activate proteins by adding a nontoxic, bioorthogonal phosphine trigger. We applied this methodology to control a post-translational modification (SUMOylation) in live cells, using native modification machinery. This work significantly improves the rate, yield, and tunability of the Staudinger reduction-based activation, paving the way for its application in other proteins and organisms.
Collapse
Affiliation(s)
- Joshua S. Wesalo
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260 (USA)
| | - Ji Luo
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260 (USA)
| | - Kunihiko Morihiro
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260 (USA)
| | - Jihe Liu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260 (USA)
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260 (USA)
| |
Collapse
|
8
|
Abstract
Covalent modification of proteins with the small ubiquitin-related modifier (SUMO) is found in all eukaryotes and is involved in many important processes. SUMO attachment may change interaction properties, subcellular localization, or stability of a modified protein. Usually, only a small fraction of a protein is modified at a given time because sumoylation is a highly dynamic process. The sumoylated state of a protein is controlled by the activity of the sumoylation enzymes that promote either their mono- or poly-sumoylation (SUMO chain formation), by SUMO proteases that reverse these modifications, and by SUMO-targeted ubiquitin ligases (STUbL, ULS) that mediate their degradation by the proteasome. While some organisms, such as humans, express multiple isoforms, budding yeast SUMO is encoded by a single and essential gene termed SMT3. The analysis of the simpler SUMO system in budding yeast has been instrumental in the identification of enzymes acting on this modification and controlling its dynamics. Sumoylation of proteins changes dramatically during the cell division cycle and under various stress conditions. Here we summarize various approaches that employ Saccharomyces cerevisiae as a model system to study the dynamics of sumoylation and how it is controlled.
Collapse
|
9
|
Li H, Zhang W, Yan M, Qiu J, Chen J, Sun X, Chen X, Song L, Zhang Y. Nucleolar and spindle associated protein 1 promotes metastasis of cervical carcinoma cells by activating Wnt/β-catenin signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:33. [PMID: 30678687 PMCID: PMC6346521 DOI: 10.1186/s13046-019-1037-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/13/2019] [Indexed: 01/06/2023]
Abstract
Background The primary obstacle to treat cervical cancer is its high prevalence of metastasis, which severely affects patients’ quality of life and survival time. Nucleolar and spindle associated protein 1 (NUSAP1) has been implicated in the development, progression, and metastasis in several types of cancer. However, its oncogenic role in cervical cancer remains unclear. Methods Western blot assay and immunohistochemistry were used to determine the expression of NUSAP1 in 21 clinical fresh Cervical cancer tissues and 233 clinicopathologically characterized cervical cancer specimens. The biological roles of NUSAP1 in the metastasis of cervical cancer were investigated both in vitro by EMT, Side population analysis and Transwell assays and so on, and in vivo using a mouse 4w model of hematogenous metastasis and lymph node metastasis. Bioinformatics analysis, luciferase reporter analysis, immunoprecipitation and immunoblotting of nuclear and cytoplasmic cellular fractions were applied to discern and examine the relationshipbetween NUSAP1 and its potential targets. Results The results demonstrated that NUSAP1 was upregulated in cervical cancer cells and tissues, correlated positively with metastasis and poor clinical outcome of patients. High expression of NUSAP1 promoted metastasis by enhancing cancer stem cell (CSC) traits and epithelial-mesenchyme transition (EMT) progression, while silencing of NUSAP1 reduced CSC traits and EMT progression. Mechanistically, upregulation of NUSAP1 induced SUMOylation of TCF4 via interacting with SUMO E3 ligase Ran-binding protein 2 (RanBP2) and hyperactivated Wnt/β-catenin signaling in cervical cancer cells. Additionally, NUSAP1-induced cervical cancer cells metastasis and the cancer stem cell phenotype were abrogated with the Wnt/β-catenin signaling inhibitor XAV-939 treatment. Importantly, co-therapy of conventional treatment and XAV-939 will provide a novel and effective treatment for NUSAP1-ovexpressed cervical cancer patients. Conclusions Our results demonstrate thatNUSAP1 upregulation contributes to metastasis of cervical cancer by promoting CSC properties and EMT via Wnt/β-catenin signaling and XAV-939 might serve as a potential tailored therapeutic option for patients with NUSAP1-ovexpressed cervical cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1037-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Han Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weijing Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ming Yan
- Department of Obstetrics Gynecology, The First Pepole's Hospital, Foshan, Guangdong, China
| | - Jiaqi Qiu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jueming Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaoying Sun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangfu Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Libing Song
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yanna Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
10
|
Steffensen AB, Andersen MN, Mutsaers N, Mujezinovic A, Schmitt N. SUMO co-expression modifies K V 11.1 channel activity. Acta Physiol (Oxf) 2018; 222. [PMID: 28888063 DOI: 10.1111/apha.12974] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 08/31/2017] [Accepted: 09/06/2017] [Indexed: 11/27/2022]
Abstract
AIM The voltage-gated potassium channel KV 11.1 is the molecular basis for the IKr current, which plays an important role in cardiac physiology. Its malfunction is associated with both inherited and acquired cardiac arrhythmias. Native currents differ from those in experimental models, suggesting additional regulatory mechanisms. We hypothesized that the post-translational modification sumoylation fine-tunes channel activity. METHODS The functional effects of sumoylation on KV 11.1 were addressed by employing two-electrode voltage-clamp (TEVC) experiments in Xenopus laevis oocytes. Site-directed mutagenesis enabled a further analysis of the SUMO-target amino acids. We assessed protein expression levels and used confocal imaging for localization studies. RESULTS Co-expression with Ubc9 and SUMO alters the electrophysiological properties of KV 11.1 leading to a decrease in steady-state current amplitude largely due to faster inactivation and alteration of deactivation kinetics. We identified three lysines (K21, K93 and K116) in the PAS domain as the putative SUMO-targets. CONCLUSION This study indicates KV 11.1 as a sumoylation target and offers three main targets: K21, K93, and K116. Furthermore, it proposes an underlying mechanism for the observed kinetic impact of the PAS domain.
Collapse
Affiliation(s)
- A. B. Steffensen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - M. N. Andersen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - N. Mutsaers
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - A. Mujezinovic
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| | - N. Schmitt
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; Danish National Research Foundation Centre for Cardiac Arrhythmia; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
11
|
Jalal D, Chalissery J, Hassan AH. Genome maintenance in Saccharomyces cerevisiae: the role of SUMO and SUMO-targeted ubiquitin ligases. Nucleic Acids Res 2017; 45:2242-2261. [PMID: 28115630 PMCID: PMC5389695 DOI: 10.1093/nar/gkw1369] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/02/2017] [Indexed: 01/08/2023] Open
Abstract
The genome of the cell is often exposed to DNA damaging agents and therefore requires an intricate well-regulated DNA damage response (DDR) to overcome its deleterious effects. The DDR needs proper regulation for its timely activation, repression, as well as appropriate choice of repair pathway. Studies in Saccharomyces cerevisiae have advanced our understanding of the DNA damage response, as well as the mechanisms the cell employs to maintain genome stability and how these mechanisms are regulated. Eukaryotic cells utilize post-translational modifications as a means for fine-tuning protein functions. Ubiquitylation and SUMOylation involve the attachment of small protein molecules onto proteins to modulate function or protein–protein interactions. SUMO in particular, was shown to act as a molecular glue when DNA damage occurs, facilitating the assembly of large protein complexes in repair foci. In other instances, SUMOylation alters a protein's biochemical activities, and interactions. SUMO-targeted ubiquitin ligases (STUbLs) are enzymes that target SUMOylated proteins for ubiquitylation and subsequent degradation, providing a function for the SUMO modification in the regulation and disassembly of repair complexes. Here, we discuss the major contributions of SUMO and STUbLs in the regulation of DNA damage repair pathways as well as in the maintenance of critical regions of the genome, namely rDNA regions, telomeres and the 2 μm circle in budding yeast.
Collapse
Affiliation(s)
- Deena Jalal
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al-Ain, UAE
| | - Jisha Chalissery
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al-Ain, UAE
| | - Ahmed H Hassan
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al-Ain, UAE
| |
Collapse
|
12
|
Dustrude ET, Perez-Miller S, François-Moutal L, Moutal A, Khanna M, Khanna R. A single structurally conserved SUMOylation site in CRMP2 controls NaV1.7 function. Channels (Austin) 2017; 11:316-328. [PMID: 28277940 DOI: 10.1080/19336950.2017.1299838] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The neuronal collapsin response mediator protein 2 (CRMP2) undergoes several posttranslational modifications that codify its functions. Most recently, CRMP2 SUMOylation (addition of small ubiquitin like modifier (SUMO)) was identified as a key regulatory step within a modification program that codes for CRMP2 interaction with, and trafficking of, voltage-gated sodium channel NaV1.7. In this paper, we illustrate the utility of combining sequence alignment within protein families with structural analysis to identify, from several putative SUMOylation sites, those that are most likely to be biologically relevant. Co-opting this principle to CRMP2, we demonstrate that, of 3 sites predicted to be SUMOylated in CRMP2, only the lysine 374 site is a SUMOylation client. A reduction in NaV1.7 currents was the corollary of the loss of CRMP2 SUMOylation at this site. A 1.78-Å-resolution crystal structure of mouse CRMP2 was solved using X-ray crystallography, revealing lysine 374 as buried within the CRMP2 tetramer interface but exposed in the monomer. Since CRMP2 SUMOylation is dependent on phosphorylation, we postulate that this state forces CRMP2 toward a monomer, exposing the SUMO site and consequently, resulting in constitutive regulation of NaV1.7.
Collapse
Affiliation(s)
- Erik Thomas Dustrude
- a Department of Pharmacology, College of Medicine , University of Arizona , Tucson , AZ , USA
| | - Samantha Perez-Miller
- a Department of Pharmacology, College of Medicine , University of Arizona , Tucson , AZ , USA
| | - Liberty François-Moutal
- a Department of Pharmacology, College of Medicine , University of Arizona , Tucson , AZ , USA
| | - Aubin Moutal
- a Department of Pharmacology, College of Medicine , University of Arizona , Tucson , AZ , USA
| | - May Khanna
- a Department of Pharmacology, College of Medicine , University of Arizona , Tucson , AZ , USA
| | - Rajesh Khanna
- a Department of Pharmacology, College of Medicine , University of Arizona , Tucson , AZ , USA.,b Department of Anesthesiology, College of Medicine , University of Arizona , Tucson , AZ , USA.,c Neuroscience Graduate Interdisciplinary Program, College of Medicine , University of Arizona , Tucson , AZ , USA
| |
Collapse
|
13
|
MxA Mediates SUMO-Induced Resistance to Vesicular Stomatitis Virus. J Virol 2016; 90:6598-6610. [PMID: 27170750 DOI: 10.1128/jvi.00722-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/03/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Multiple cellular pathways are regulated by small ubiquitin-like modifier (SUMO) modification, including ubiquitin-mediated proteolysis, signal transduction, innate immunity, and antiviral defense. In the study described in this report, we investigated the effects of SUMO on the replication of two members of the Rhabdoviridae family, vesicular stomatitis virus (VSV) and rabies virus (RABV). We show that stable expression of SUMO in human cells confers resistance to VSV infection in an interferon-independent manner. We demonstrate that SUMO expression did not alter VSV entry but blocked primary mRNA synthesis, leading to a reduction of viral protein synthesis and viral production, thus protecting cells from VSV-induced cell lysis. MxA is known to inhibit VSV primary transcription. Interestingly, we found that the MxA protein was highly stabilized in SUMO-expressing cells. Furthermore, extracts from cells stably expressing SUMO exhibited an increase in MxA oligomers, suggesting that SUMO plays a role in protecting MxA from degradation, thus providing a stable intracellular pool of MxA available to combat invading viruses. Importantly, MxA depletion in SUMO-expressing cells abrogated the anti-VSV effect of SUMO. Furthermore, SUMO expression resulted in interferon-regulatory factor 3 (IRF3) SUMOylation, subsequently decreasing RABV-induced IRF3 phosphorylation and interferon synthesis. As expected, this rendered SUMO-expressing cells more sensitive to RABV infection, even though MxA was stabilized in SUMO-expressing cells, since its expression did not confer resistance to RABV. Our findings demonstrate opposing effects of SUMO expression on two viruses of the same family, intrinsically inhibiting VSV infection through MxA stabilization while enhancing RABV infection by decreasing IFN induction. IMPORTANCE We report that SUMO expression reduces interferon synthesis upon RABV or VSV infection. Therefore, SUMO renders cells more sensitive to RABV but unexpectedly renders cells resistant to VSV by blocking primary mRNA synthesis. Unlike the interferon-mediated innate immune response, intrinsic antiviral resistance is mediated by constitutively expressed restriction factors. Among the various anti-VSV restriction factors, only MxA is known to inhibit VSV primary transcription, and we show here that its expression does not alter RABV infection. Interestingly, MxA depletion abolished the inhibition of VSV by SUMO, demonstrating that MxA mediates SUMO-induced intrinsic VSV resistance. Furthermore, MxA oligomerization is known to be critical for its protein stability, and we show that higher levels of oligomers were formed in cells expressing SUMO than in wild-type cells, suggesting that SUMO may play a role in protecting MxA from degradation, providing a stable intracellular pool of MxA able to protect cells from viral infection.
Collapse
|
14
|
Puntambekar SS, Nyayanit D, Saxena P, Gadgil CJ. Identification of Unintuitive Features of Sumoylation through Mathematical Modeling. J Biol Chem 2016; 291:9458-68. [PMID: 26861881 PMCID: PMC4850286 DOI: 10.1074/jbc.m115.676122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 01/04/2016] [Indexed: 11/06/2022] Open
Abstract
Sumoylation is a multistep, multienzymatic post-translational modification in which a small ubiquitin-like modifier protein (SUMO) is attached to the target. We present the first mathematical model for sumoylation including enzyme mechanism details such as autosumoylation of E2 and multifunctional nature of SENP. Simulations and analysis reveal three nonobvious properties for the long term response, modeled as an open system: (i) the steady state sumoylation level is robust to variation in several enzyme properties; (ii) even when autosumoylation of E2 results in equal or higher activity, the target sumoylation levels are lower; and (iii) there is an optimal SENP concentration at which steady state target sumoylation level is maximum. These results are qualitatively different for a short term response modeled as a closed system, where e.g. sumoylation always decreases with increasing SENP levels. Simulations with multiple targets suggest that the available SUMO is limiting, indicating a possible explanation for the experimentally observed low fractional sumoylation. We predict qualitative differences in system responses at short post-translational and longer transcriptional time scales. We thus use this mechanism-based model to explain system properties and generate testable hypotheses for existence and mechanism of unexpected responses.
Collapse
Affiliation(s)
- Shraddha S Puntambekar
- From the Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune-411008, and the Academy of Scientific and Innovative Research (AcSIR), CSIR-NCL Campus, Pune-411008, and
| | - Dimpal Nyayanit
- From the Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune-411008, and the Academy of Scientific and Innovative Research (AcSIR), CSIR-NCL Campus, Pune-411008, and
| | - Priyanka Saxena
- the Academy of Scientific and Innovative Research (AcSIR), CSIR-NCL Campus, Pune-411008, and the CSIR-Institute of Genomics and Integrative Biology, New Delhi 110020, India
| | - Chetan J Gadgil
- From the Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune-411008, and the Academy of Scientific and Innovative Research (AcSIR), CSIR-NCL Campus, Pune-411008, and the CSIR-Institute of Genomics and Integrative Biology, New Delhi 110020, India
| |
Collapse
|
15
|
Hannoun Z, Maarifi G, Chelbi-Alix MK. The implication of SUMO in intrinsic and innate immunity. Cytokine Growth Factor Rev 2016; 29:3-16. [PMID: 27157810 DOI: 10.1016/j.cytogfr.2016.04.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/31/2022]
Abstract
Since its discovery, SUMOylation has emerged as a key post-translational modification involved in the regulation of host-virus interactions. SUMOylation has been associated with the replication of a large number of viruses, either through the direct modification of viral proteins or through the modulation of cellular proteins implicated in antiviral defense. SUMO can affect protein function via covalent or non-covalent binding. There is growing evidence that SUMO regulates several host proteins involved in intrinsic and innate immunity, thereby contributing to the process governing interferon production during viral infection; as well as the interferon-activated Jak/STAT pathway. Unlike the interferon-mediated innate immune response, intrinsic antiviral resistance is mediated by constitutively expressed antiviral proteins (defined as restriction factors), which confer direct viral resistance through a variety of mechanisms. The aim of this review is to evaluate the role of SUMO in intrinsic and innate immunity; highlighting the involvement of the TRIM family proteins, with a specific focus on the mechanism through which SUMO affects i- interferon production upon viral infection, ii-interferon Jak/STAT signaling and biological responses, iii-the relationship between restriction factors and RNA viruses.
Collapse
Affiliation(s)
- Zara Hannoun
- INSERM UMR-S 1124, Université Paris Descartes, Paris, France
| | | | | |
Collapse
|
16
|
Ohkuni K, Takahashi Y, Fulp A, Lawrimore J, Au WC, Pasupala N, Levy-Myers R, Warren J, Strunnikov A, Baker RE, Kerscher O, Bloom K, Basrai MA. SUMO-Targeted Ubiquitin Ligase (STUbL) Slx5 regulates proteolysis of centromeric histone H3 variant Cse4 and prevents its mislocalization to euchromatin. Mol Biol Cell 2016; 27:mbc.E15-12-0827. [PMID: 26960795 PMCID: PMC4850037 DOI: 10.1091/mbc.e15-12-0827] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/17/2016] [Accepted: 03/03/2016] [Indexed: 12/14/2022] Open
Abstract
Centromeric histone H3, CENP-ACse4, is essential for faithful chromosome segregation. Stringent regulation of cellular levels of CENP-ACse4 restricts its localization to centromeres. Mislocalization of CENP-ACse4 is associated with aneuploidy in yeast, flies and tumorigenesis in human cells; thus, defining pathways that regulate CENP-A levels is critical for understanding how mislocalization of CENP-A contributes to aneuploidy in human cancers. Previous work in budding yeast has shown that ubiquitination of overexpressed Cse4 by Psh1, an E3 ligase, partially contributes to proteolysis of Cse4. Here, we provide the first evidence that Cse4 is sumoylated by E3 ligases Siz1 and Siz2 in vivo and in vitro. Ubiquitination of Cse4 by Small Ubiquitin-related Modifier (SUMO)-Targeted Ubiquitin Ligase (STUbL) Slx5 plays a critical role in proteolysis of Cse4 and prevents mislocalization of Cse4 to euchromatin under normal physiological conditions. Accumulation of sumoylated Cse4 species and increased stability of Cse4 in slx5∆ strains suggest that sumoylation precedes ubiquitin-mediated proteolysis of Cse4. Slx5-mediated Cse4 proteolysis is independent of Psh1 since slx5∆ psh1∆ strains exhibit higher levels of Cse4 stability and mislocalization compared to either slx5∆ or psh1∆ strains. Our results demonstrate a role for Slx5 in ubiquitin-mediated proteolysis of Cse4 to prevent its mislocalization and maintain genome stability.
Collapse
Affiliation(s)
- Kentaro Ohkuni
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yoshimitsu Takahashi
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Alyona Fulp
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Josh Lawrimore
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Wei-Chun Au
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Nagesh Pasupala
- Biology Department, The College of William & Mary, Williamsburg, VA 23187
| | - Reuben Levy-Myers
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
- Biology Department, The College of William & Mary, Williamsburg, VA 23187
| | - Jack Warren
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Richard E. Baker
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Oliver Kerscher
- Biology Department, The College of William & Mary, Williamsburg, VA 23187
| | - Kerry Bloom
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Munira A. Basrai
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
17
|
Tang X, Li W, Xing J, Sheng X, Zhan W. SUMO and SUMO-Conjugating Enzyme E2 UBC9 Are Involved in White Spot Syndrome Virus Infection in Fenneropenaeus chinensis. PLoS One 2016; 11:e0150324. [PMID: 26927328 PMCID: PMC4771164 DOI: 10.1371/journal.pone.0150324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/11/2016] [Indexed: 01/18/2023] Open
Abstract
In previous work, small ubiquitin-like modifier (SUMO) in hemocytes of Chinese shrimp Fenneropenaeus chinensis was found to be up-regulated post-white spot syndrome virus (WSSV) infection using proteomic approach. However, the role of SUMO in viral infection is still unclear. In the present work, full length cDNAs of SUMO (FcSUMO) and SUMO-conjugating enzyme E2 UBC9 (FcUBC9) were cloned from F. chinensis using rapid amplification of cDNA ends approach. The open reading frame (ORF) of FcSUMO encoded a 93 amino acids peptide with the predicted molecular weight (M.W) of 10.55 kDa, and the UBC9 ORF encoded a 160 amino acids peptide with the predicted M.W of 18.35 kDa. By quantitative real-time RT-PCR, higher mRNA transcription levels of FcSUMO and FcUBC9 were detected in hemocytes and ovary of F. chinensis, and the two genes were significantly up-regulated post WSSV infection. Subsequently, the recombinant proteins of FcSUMO and FcUBC9 were expressed in Escherichia coli BL21 (DE3), and employed as immunogens for the production of polyclonal antibody (PAb). Indirect immunofluorescence assay revealed that the FcSUMO and UBC9 proteins were mainly located in the hemocytes nuclei. By western blotting, a 13.5 kDa protein and a 18.7 kDa protein in hemocytes were recognized by the PAb against SUMO or UBC9 respectively. Furthermore, gene silencing of FcSUMO and FcUBC9 were performed using RNA interference, and the results showed that the number of WSSV copies and the viral gene expressions were inhibited by knockdown of either SUMO or UBC9, and the mortalities of shrimp were also reduced. These results indicated that FcSUMO and FcUBC9 played important roles in WSSV infection.
Collapse
Affiliation(s)
- Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, Yushan road 5, Qingdao, 266003, PR China
| | - Wei Li
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, Yushan road 5, Qingdao, 266003, PR China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, Yushan road 5, Qingdao, 266003, PR China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, Yushan road 5, Qingdao, 266003, PR China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, Yushan road 5, Qingdao, 266003, PR China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Aoshanwei Town, Jimo, Qingdao, 266071, China
- * E-mail:
| |
Collapse
|
18
|
Extraembryonic but not embryonic SUMO-specific protease 2 is required for heart development. Sci Rep 2016; 6:20999. [PMID: 26883797 PMCID: PMC4756675 DOI: 10.1038/srep20999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/15/2016] [Indexed: 02/01/2023] Open
Abstract
SUMO-specific protease 2 (SENP2) activities to remove SUMO from its substrates is essential for development of trophoblast stem cells, niches and lineages. Global deletion of SENP2 leads to midgestation lethality, and causes severe defects in the placenta which is accompanied by embryonic brain and heart abnormalities. Because of the placental deficiencies, the role of SENP2 in development of the embryonic tissues has not been properly determined. The brain and heart abnormalities may be secondary to placental insufficiency. Here we have created a new mouse strain permitting conditional inactivation of SENP2. Mice homozygous for germline deletion of the conditional allele exhibit trophoblast defects and embryonic abnormalities resembling the global SENP2 knockout. However, tissue-specific disruptions of SENP2 demonstrate its dispensable role in embryogenesis. Placental expression of SENP2 is necessary and sufficient for embryonic heart and brain development. Using a protease deficient model, we further demonstrate the requirement of SENP2-dependent SUMO modification in development of all major trophoblast lineages. SENP2 regulates sumoylation of Mdm2 which controls p53 activities critical for G-S transition of mitotic division and endoreduplication in trophoblast proliferation and differentiation, respectively. The differentiation of trophoblasts is also dependent on SENP2-mediated activation of p57Kip2, a CDK-specific inhibitor required for endoreduplication.
Collapse
|
19
|
Mas A, Amenós M, Lois LM. Quantitative Analysis of Subcellular Distribution of the SUMO Conjugation System by Confocal Microscopy Imaging. Methods Mol Biol 2016; 1450:135-150. [PMID: 27424751 DOI: 10.1007/978-1-4939-3759-2_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Different studies point to an enrichment in SUMO conjugation in the cell nucleus, although non-nuclear SUMO targets also exist. In general, the study of subcellular localization of proteins is essential for understanding their function within a cell. Fluorescence microscopy is a powerful tool for studying subcellular protein partitioning in living cells, since fluorescent proteins can be fused to proteins of interest to determine their localization. Subcellular distribution of proteins can be influenced by binding to other biomolecules and by posttranslational modifications. Sometimes these changes affect only a portion of the protein pool or have a partial effect, and a quantitative evaluation of fluorescence images is required to identify protein redistribution among subcellular compartments. In order to obtain accurate data about the relative subcellular distribution of SUMO conjugation machinery members, and to identify the molecular determinants involved in their localization, we have applied quantitative confocal microscopy imaging. In this chapter, we will describe the fluorescent protein fusions used in these experiments, and how to measure, evaluate, and compare average fluorescence intensities in cellular compartments by image-based analysis. We show the distribution of some components of the Arabidopsis SUMOylation machinery in epidermal onion cells and how they change their distribution in the presence of interacting partners or even when its activity is affected.
Collapse
Affiliation(s)
- Abraham Mas
- Development Program, CRAG (CSIC-IRTA-UAB-UB), Edifici CRAG-Campus UAB, Bellaterra (Cerdanyola del Vallés), 08193, Barcelona, Spain
| | - Montse Amenós
- Confocal Microscopy Facility, CRAG (CSIC-IRTA-UAB-UB), Edifici CRAG-Campus UAB, Bellaterra (Cerdanyola del Vallés), 08193, Barcelona, Spain
| | - L Maria Lois
- Development Program, CRAG (CSIC-IRTA-UAB-UB), Edifici CRAG-Campus UAB, Bellaterra (Cerdanyola del Vallés), 08193, Barcelona, Spain.
| |
Collapse
|
20
|
Depletion of UBC9 Causes Nuclear Defects during the Vegetative and Sexual Life Cycles in Tetrahymena thermophila. EUKARYOTIC CELL 2015; 14:1240-52. [PMID: 26453653 DOI: 10.1128/ec.00115-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/03/2015] [Indexed: 11/20/2022]
Abstract
Ubc9p is the sole E2-conjugating enzyme for SUMOylation, and its proper function is required for regulating key nuclear events such as transcription, DNA repair, and mitosis. In Tetrahymena thermophila, the genome is separated into a diploid germ line micronucleus (MIC) that divides by mitosis and a polyploid somatic macronucleus (MAC) that divides amitotically. This unusual nuclear organization provides novel opportunities for the study of SUMOylation and Ubc9p function. We identified the UBC9 gene and demonstrated that its complete deletion from both MIC and MAC genomes is lethal. Rescue of the lethal phenotype with a GFP-UBC9 fusion gene driven by a metallothionein promoter generated a cell line with CdCl2-dependent expression of green fluorescent protein (GFP)-Ubc9p. Depletion of Ubc9p in vegetative cells resulted in the loss of MICs, but MACs continued to divide. In contrast, expression of catalytically inactive Ubc9p resulted in the accumulation of multiple MICs. Critical roles for Ubc9p were also identified during the sexual life cycle of Tetrahymena. Cell lines that were depleted for Ubc9p did not form mating pairs and therefore could not complete any of the subsequent stages of conjugation, including meiosis and macronuclear development. Mating between cells expressing catalytically inactive Ubc9p resulted in arrest during macronuclear development, consistent with our observation that Ubc9p accumulates in the developing macronucleus.
Collapse
|
21
|
Alonso A, Greenlee M, Matts J, Kline J, Davis KJ, Miller RK. Emerging roles of sumoylation in the regulation of actin, microtubules, intermediate filaments, and septins. Cytoskeleton (Hoboken) 2015; 72:305-39. [PMID: 26033929 PMCID: PMC5049490 DOI: 10.1002/cm.21226] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 12/29/2022]
Abstract
Sumoylation is a powerful regulatory system that controls many of the critical processes in the cell, including DNA repair, transcriptional regulation, nuclear transport, and DNA replication. Recently, new functions for SUMO have begun to emerge. SUMO is covalently attached to components of each of the four major cytoskeletal networks, including microtubule-associated proteins, septins, and intermediate filaments, in addition to nuclear actin and actin-regulatory proteins. However, knowledge of the mechanisms by which this signal transduction system controls the cytoskeleton is still in its infancy. One story that is beginning to unfold is that SUMO may regulate the microtubule motor protein dynein by modification of its adaptor Lis1. In other instances, cytoskeletal elements can both bind to SUMO non-covalently and also be conjugated by it. The molecular mechanisms for many of these new functions are not yet clear, but are under active investigation. One emerging model links the function of MAP sumoylation to protein degradation through SUMO-targeted ubiquitin ligases, also known as STUbL enzymes. Other possible functions for cytoskeletal sumoylation are also discussed.
Collapse
Affiliation(s)
- Annabel Alonso
- Department of Biochemistry and Molecular BiologyOklahoma State UniversityStillwaterOklahoma
| | - Matt Greenlee
- Department of Biochemistry and Molecular BiologyOklahoma State UniversityStillwaterOklahoma
| | - Jessica Matts
- Department of Biochemistry and Molecular BiologyOklahoma State UniversityStillwaterOklahoma
| | - Jake Kline
- Department of Biochemistry and Molecular BiologyOklahoma State UniversityStillwaterOklahoma
| | - Kayla J. Davis
- Department of Biochemistry and Molecular BiologyOklahoma State UniversityStillwaterOklahoma
| | - Rita K. Miller
- Department of Biochemistry and Molecular BiologyOklahoma State UniversityStillwaterOklahoma
| |
Collapse
|
22
|
Alontaga AY, Ambaye ND, Li YJ, Vega R, Chen CH, Bzymek KP, Williams JC, Hu W, Chen Y. RWD Domain as an E2 (Ubc9)-Interaction Module. J Biol Chem 2015; 290:16550-9. [PMID: 25918163 DOI: 10.1074/jbc.m115.644047] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Indexed: 11/06/2022] Open
Abstract
An RWD domain is a well conserved domain found through bioinformatic analysis of the human proteome sequence; however, its function has been unknown. Ubiquitin-like modifications require the catalysis of three enzymes generally known as E1, E2, and E3. We solved the crystal structure of the E2 for the small ubiquitin-like modifiers (SUMO) in complex with an RWD domain and confirmed the structure using solution NMR analysis. The binding surface of RWD on Ubc9 is located near the N terminus of Ubc9 that is known to be involved in noncovalent binding of the proteins in the conjugation machinery, including a domain of E1, SUMO, and an E3 ligase. NMR data indicate that the RWD domain does not bind to SUMO and E1. The interaction between RWD and Ubc9 has a Kd of 32 ± 4 μM. Consistent with the structure and binding affinity and in contrast to a previous report, the RWD domain and RWDD3 have minimal effects on global SUMOylation. The structural and biochemical information presented here forms the basis for further investigation of the functions of RWD-containing proteins.
Collapse
Affiliation(s)
| | | | - Yi-Jia Li
- From the Department of Molecular Medicine and
| | - Ramir Vega
- From the Department of Molecular Medicine and
| | | | | | | | - Weidong Hu
- the NMR Core Facility, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - Yuan Chen
- From the Department of Molecular Medicine and
| |
Collapse
|
23
|
Venegas-Vega C, Nieto-Martínez K, Martínez-Herrera A, Gómez-Laguna L, Berumen J, Cervantes A, Kofman S, Fernández-Ramírez F. 19q13.11 microdeletion concomitant with ins(2;19)(p25.3;q13.1q13.4)dn in a boy: potential role of UBA2 in the associated phenotype. Mol Cytogenet 2014; 7:61. [PMID: 25516771 PMCID: PMC4266984 DOI: 10.1186/s13039-014-0061-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 08/26/2014] [Indexed: 01/17/2023] Open
Abstract
The 19q13.11 microdeletion syndrome (MIM613026) is a clinically recognisable condition in which a 324-kb minimal overlapping critical region has been recently described. However, genes not included within this region, such as WTIP and UBA2, have been proposed to contribute to the clinical characteristics observed in patients. Using cytogenetic techniques, single nucleotide polymorphism arrays, and the quantitative polymerase chain reaction, we identified a novel case with a 2.49-Mb deletion derived from a de novo chromosomal rearrangement. Based on a review of the literature, we support the notion that UBA2 haploinsufficiency could contribute to the phenotype of this rare genomic disorder. UBA2 belongs to a protein complex with sumoylation activity, and several transcription factors, hormone receptors, and signalling proteins related to brain and sexual development are regulated by this post-translational modification. Additional clinical reports and further research on UBA2 molecular function are warranted.
Collapse
Affiliation(s)
- Carlos Venegas-Vega
- Unidad de Genética, Hospital General de México, Dr. Balmis 148, México, D.F 06726 México ; Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México, D.F 04510 México
| | - Karem Nieto-Martínez
- Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México, D.F 04510 México
| | - Alejandro Martínez-Herrera
- Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México, D.F 04510 México
| | - Laura Gómez-Laguna
- Unidad de Genética, Hospital General de México, Dr. Balmis 148, México, D.F 06726 México
| | - Jaime Berumen
- Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México, D.F 04510 México ; Unidad de Medicina Genómica, Hospital General de México, Dr. Balmis 148, México, D.F 06726 México
| | - Alicia Cervantes
- Unidad de Genética, Hospital General de México, Dr. Balmis 148, México, D.F 06726 México ; Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México, D.F 04510 México
| | - Susana Kofman
- Unidad de Genética, Hospital General de México, Dr. Balmis 148, México, D.F 06726 México ; Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, México, D.F 04510 México
| | | |
Collapse
|
24
|
Watts FZ, Baldock R, Jongjitwimol J, Morley SJ. Weighing up the possibilities: Controlling translation by ubiquitylation and sumoylation. ACTA ACUST UNITED AC 2014; 2:e959366. [PMID: 26779408 DOI: 10.4161/2169074x.2014.959366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/01/2014] [Accepted: 05/12/2014] [Indexed: 12/15/2022]
Abstract
Regulation of protein synthesis is of fundamental importance to cells. It has a critical role in the control of gene expression, and consequently cell growth and proliferation. The importance of this control is supported by the fact that protein synthesis is frequently upregulated in tumor cells. The major point at which regulation occurs is the initiation stage. Initiation of translation involves the interaction of several proteins to form the eIF4F complex, the recognition of the mRNA by this complex, and the subsequent recruitment of the 40S ribosomal subunit to the mRNA. This results in the formation of the 48S complex that then scans the mRNA for the start codon, engages the methionyl-tRNA and eventually forms the mature 80S ribosome which is elongation-competent. Formation of the 48S complex is regulated by the availability of individual initiation factors and through specific protein-protein interactions. Both of these events can be regulated by post-translational modification by ubiquitin or Ubls (ubiquitin-like modifiers) such as SUMO or ISG15. We provide here a summary of translation initiation factors that are modified by ubiquitin or Ubls and, where they have been studied in detail, describe the role of these modifications and their effects on regulating protein synthesis.
Collapse
Affiliation(s)
- Felicity Z Watts
- Genome Damage and Stability Center; School of Life Sciences; University of Sussex ; Falmer, Brighton, UK
| | - Robert Baldock
- Genome Damage and Stability Center; School of Life Sciences; University of Sussex ; Falmer, Brighton, UK
| | - Jirapas Jongjitwimol
- Genome Damage and Stability Center; School of Life Sciences; University of Sussex ; Falmer, Brighton, UK
| | - Simon J Morley
- Department of Biochemistry and Biomedical Science; School of Life Sciences; University of Sussex ; Brighton, UK
| |
Collapse
|
25
|
Disruption of SUMO-specific protease 2 induces mitochondria mediated neurodegeneration. PLoS Genet 2014; 10:e1004579. [PMID: 25299344 PMCID: PMC4191884 DOI: 10.1371/journal.pgen.1004579] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
Post-translational modification of proteins by small ubiquitin-related modifier (SUMO) is reversible and highly evolutionarily conserved from yeasts to humans. Unlike ubiquitination with a well-established role in protein degradation, sumoylation may alter protein function, activity, stability and subcellular localization. Members of SUMO-specific protease (SENP) family, capable of SUMO removal, are involved in the reversed conjugation process. Although SUMO-specific proteases are known to reverse sumoylation in many well-defined systems, their importance in mammalian development and pathogenesis remains largely elusive. In patients with neurodegenerative diseases, aberrant accumulation of SUMO-conjugated proteins has been widely described. Several aggregation-prone proteins modulated by SUMO have been implicated in neurodegeneration, but there is no evidence supporting a direct involvement of SUMO modification enzymes in human diseases. Here we show that mice with neural-specific disruption of SENP2 develop movement difficulties which ultimately results in paralysis. The disruption induces neurodegeneration where mitochondrial dynamics is dysregulated. SENP2 regulates Drp1 sumoylation and stability critical for mitochondrial morphogenesis in an isoform-specific manner. Although dispensable for development of neural cell types, this regulatory mechanism is necessary for their survival. Our findings provide a causal link of SUMO modification enzymes to apoptosis of neural cells, suggesting a new pathogenic mechanism for neurodegeneration. Exploring the protective effect of SENP2 on neuronal cell death may uncover important preventive and therapeutic strategies for neurodegenerative diseases.
Collapse
|
26
|
Park YS, Kang JW, Lee DH, Kim MS, Bak Y, Yang Y, Lee HG, Hong J, Yoon DY. Interleukin-32α modulates promyelocytic leukemia zinc finger gene activity by inhibiting protein kinase Cɛ-dependent sumoylation. Int J Biochem Cell Biol 2014; 55:136-43. [DOI: 10.1016/j.biocel.2014.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 07/17/2014] [Accepted: 08/21/2014] [Indexed: 01/20/2023]
|
27
|
Sarangi P, Altmannova V, Holland C, Bartosova Z, Hao F, Anrather D, Ammerer G, Lee SE, Krejci L, Zhao X. A versatile scaffold contributes to damage survival via sumoylation and nuclease interactions. Cell Rep 2014; 9:143-152. [PMID: 25263559 PMCID: PMC4280569 DOI: 10.1016/j.celrep.2014.08.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/23/2014] [Accepted: 08/22/2014] [Indexed: 11/30/2022] Open
Abstract
DNA repair scaffolds mediate specific DNA and protein interactions in order to assist repair enzymes in recognizing and removing damaged sequences. Many scaffold proteins are dedicated to repairing a particular type of lesion. Here, we show that the budding yeast Saw1 scaffold is more versatile. It helps cells cope with base lesions and protein-DNA adducts through its known function of recruiting the Rad1-Rad10 nuclease to DNA. In addition, it promotes UV survival via a mechanism mediated by its sumoylation. Saw1 sumoylation favors its interaction with another nuclease Slx1-Slx4, and this SUMO-mediated role is genetically separable from two main UV lesion repair processes. These effects of Saw1 and its sumoylation suggest that Saw1 is a multifunctional scaffold that can facilitate diverse types of DNA repair through its modification and nuclease interactions.
Collapse
Affiliation(s)
- Prabha Sarangi
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Programs in Biochemistry, Cell, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | | | - Cory Holland
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Zdenka Bartosova
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
| | - Fanfan Hao
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Dorothea Anrather
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna 1030, Austria
| | - Gustav Ammerer
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Vienna 1030, Austria
| | - Sang Eun Lee
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Division of Radiation Biology, Department of Radiation Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lumir Krejci
- Department of Biology, Masaryk University, Brno 62500, Czech Republic; National Centre for Biomolecular Research, Masaryk University, Brno 62500, Czech Republic; International Clinical Research Center, St. Anne's University Hospital in Brno, Brno 60200, Czech Republic.
| | - Xiaolan Zhao
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Programs in Biochemistry, Cell, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA.
| |
Collapse
|
28
|
Sarangi P, Bartosova Z, Altmannova V, Holland C, Chavdarova M, Lee SE, Krejci L, Zhao X. Sumoylation of the Rad1 nuclease promotes DNA repair and regulates its DNA association. Nucleic Acids Res 2014; 42:6393-404. [PMID: 24753409 PMCID: PMC4041466 DOI: 10.1093/nar/gku300] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The Saccharomyces cerevisiae Rad1-Rad10 complex is a conserved, structure-specific endonuclease important for repairing multiple types of DNA lesions. Upon recruitment to lesion sites, Rad1-Rad10 removes damaged sequences, enabling subsequent gap filling and ligation. Acting at mid-steps of repair, the association and dissociation of Rad1-Rad10 with DNA can influence repair efficiency. We show that genotoxin-enhanced Rad1 sumoylation occurs after the nuclease is recruited to lesion sites. A single lysine outside Rad1's nuclease and Rad10-binding domains is sumoylated in vivo and in vitro. Mutation of this site to arginine abolishes Rad1 sumoylation and impairs Rad1-mediated repair at high doses of DNA damage, but sustains the repair of a single double-stranded break. The timing of Rad1 sumoylation and the phenotype bias toward high lesion loads point to a post-incision role for sumoylation, possibly affecting Rad1 dissociation from DNA. Indeed, biochemical examination shows that sumoylation of Rad1 decreases the complex's affinity for DNA without affecting other protein properties. These findings suggest a model whereby sumoylation of Rad1 promotes its disengagement from DNA after nuclease cleavage, allowing it to efficiently attend to large numbers of DNA lesions.
Collapse
Affiliation(s)
- Prabha Sarangi
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA Programs in Biochemistry, Cell, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Zdenka Bartosova
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
| | | | - Cory Holland
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Melita Chavdarova
- National Centre for Biomolecular Research, Masaryk University, Brno 62500, Czech Republic
| | - Sang Eun Lee
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA Division of Radiation Biology, Department of Radiation Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lumir Krejci
- Department of Biology, Masaryk University, Brno 62500, Czech Republic National Centre for Biomolecular Research, Masaryk University, Brno 62500, Czech Republic International Clinical Research Center, St. Anne's University Hospital in Brno, Brno 60200, Czech Republic
| | - Xiaolan Zhao
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
29
|
Asymmetric Hsp90 N domain SUMOylation recruits Aha1 and ATP-competitive inhibitors. Mol Cell 2014; 53:317-29. [PMID: 24462205 DOI: 10.1016/j.molcel.2013.12.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/11/2013] [Accepted: 12/04/2013] [Indexed: 11/23/2022]
Abstract
The stability and activity of numerous signaling proteins in both normal and cancer cells depends on the dimeric molecular chaperone heat shock protein 90 (Hsp90). Hsp90's function is coupled to ATP binding and hydrolysis and requires a series of conformational changes that are regulated by cochaperones and numerous posttranslational modifications (PTMs). SUMOylation is one of the least-understood Hsp90 PTMs. Here, we show that asymmetric SUMOylation of a conserved lysine residue in the N domain of both yeast (K178) and human (K191) Hsp90 facilitates both recruitment of the adenosine triphosphatase (ATPase)-activating cochaperone Aha1 and, unexpectedly, the binding of Hsp90 inhibitors, suggesting that these drugs associate preferentially with Hsp90 proteins that are actively engaged in the chaperone cycle. Importantly, cellular transformation is accompanied by elevated steady-state N domain SUMOylation, and increased Hsp90 SUMOylation sensitizes yeast and mammalian cells to Hsp90 inhibitors, providing a mechanism to explain the sensitivity of cancer cells to these drugs.
Collapse
|
30
|
Evidence Implicating CCNB1IP1, a RING Domain-Containing Protein Required for Meiotic Crossing Over in Mice, as an E3 SUMO Ligase. Genes (Basel) 2014; 1:440-51. [PMID: 21779533 PMCID: PMC3139512 DOI: 10.3390/genes1030440] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The RING domain-containing protein CCNB1IP1 (Cyclin B1 Interacting Protein 1) is a putative ubiquitin E3 ligase that is essential for chiasmata formation, and hence fertility, in mice. Previous studies in cultured cells indicated that CCNB1IP1 targets Cyclin B for degradation, thus playing a role in cell cycle regulation. Mice homozygous for a mutant allele (mei4) of Ccnb1ip1 display no detectable phenotype other than meiotic failure from an absence of chiasmata. CCNB1IP1 is not conserved in key model organisms such as yeast and Drosophila, and there are no features of the protein that implicate clear mechanisms for a role in recombination. To gain insight into CCNB1IP1’s function in meiotic cells, we raised a specific antibody and determined that the protein appears in pachynema. This indicates that CCNB1IP1 is involved with crossover intermediate maturation, rather than early (leptotene) specification of a subset of SPO11-induced double strand breaks towards the crossover pathway. Additionally, a yeast 2-hybrid (Y2H) screen revealed that CCNB1IP1 interacts with SUMO2 and a set of proteins enriched for consensus sumoylation sites. The Y2H studies, combined with scrutiny of CCNB1IP1 domains, implicate this protein as an E3 ligase of the sumoylation cascade. We hypothesize CCNB1IP1 represents a novel meiosis-specific SUMO E3 ligase critical to resolution of recombination intermediates into mature chiasmata.
Collapse
|
31
|
Kim YS, Keyser SGL, Schneekloth JS. Synthesis of 2',3',4'-trihydroxyflavone (2-D08), an inhibitor of protein sumoylation. Bioorg Med Chem Lett 2014; 24:1094-7. [PMID: 24468414 DOI: 10.1016/j.bmcl.2014.01.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 01/02/2014] [Accepted: 01/06/2014] [Indexed: 01/22/2023]
Abstract
Protein sumoylation is a dynamic posttranslational modification involved in diverse biological processes during cellular homeostasis and development. Recently sumoylation has been shown to play a critical role in cancer, although to date there are few small molecule probes available to inhibit enzymes involved in the SUMO conjugation process. As part of a program to identify and study inhibitors of sumoylation we recently reported the discovery that 2',3',4'-trihydroxyflavone (2-D08) is a cell permeable, mechanistically unique inhibitor of protein sumoylation. The work reported herein describes an efficient synthesis of 2-D08 as well as a structurally related but inactive isomer. We also report an unanticipated Wessely-Moser rearrangement that occurs under vigorous methyl ether deprotection conditions. This rearrangement likely gave rise to 2-D08 during a deprotection step, resulting in 2-D08 appearing as a contaminant in a screening well from a commercial supplier.
Collapse
Affiliation(s)
- Yeong Sang Kim
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, 376 Boyles St., Frederick, MD 21702, USA
| | - Samantha G L Keyser
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, 376 Boyles St., Frederick, MD 21702, USA
| | - John S Schneekloth
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, 376 Boyles St., Frederick, MD 21702, USA.
| |
Collapse
|
32
|
Jentsch S, Psakhye I. Control of Nuclear Activities by Substrate-Selective and Protein-Group SUMOylation. Annu Rev Genet 2013; 47:167-86. [DOI: 10.1146/annurev-genet-111212-133453] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Stefan Jentsch
- Department of Molecular Cell Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany;
| | - Ivan Psakhye
- Department of Molecular Cell Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany;
| |
Collapse
|
33
|
Seliga J, Bielska K, Wieczorek E, Orłowski M, Niedenthal R, Ożyhar A. Multidomain sumoylation of the ecdysone receptor (EcR) from Drosophila melanogaster. J Steroid Biochem Mol Biol 2013; 138:162-73. [PMID: 23727127 DOI: 10.1016/j.jsbmb.2013.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 05/16/2013] [Accepted: 05/18/2013] [Indexed: 02/05/2023]
Abstract
The 20-hydroxyecdysone receptor (EcR) is a transcription factor belonging to the nuclear receptor superfamily. Together with the ultraspiracle nuclear receptor (Usp) it coordinates critical biological processes in insects such as development and reproduction. EcR and its ligands are used in commercially available ecdysone-inducible expression systems and are considered to be artificial gene switches with potential therapeutic applications. However, the regulation of EcR action is still unclear, especially in mammals and as far as posttranslational modifications are concerned. Up until now, there has been no study on EcR sumoylation. Using bioinformatic predictors, a Ubc9 fusion-directed sumoylation system and mutagenesis experiments, we present EcR as a new target of SUMO1 and SUMO3 modification. Our research revealed that EcR undergoes isoform-specific multisumoylation. The pattern of modification remains unchanged in the presence of the ligand and the dimerization partner. The SUMO acceptor sites are located in the DNA-binding domain and the ligand-binding domain that both exhibit structural plasticity. We also demonstrated the existence of a sumoylation site in the F region and EcRA-A/B region, both revealing characteristics of intrinsically disordered regions. The consequences of modification and the resulting impact on conformation and function may be especially crucial for the disordered sequences in these two areas. The isoform-specificity of sumoylation may explain the differences in the transcriptional activity of EcR isoforms.
Collapse
Affiliation(s)
- Justyna Seliga
- Department of Biochemistry, Faculty of Chemistry, Wrocław University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | | | | | | | | | | |
Collapse
|
34
|
Kim YS, Nagy K, Keyser S, Schneekloth JS. An electrophoretic mobility shift assay identifies a mechanistically unique inhibitor of protein sumoylation. ACTA ACUST UNITED AC 2013; 20:604-13. [PMID: 23601649 DOI: 10.1016/j.chembiol.2013.04.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/29/2013] [Accepted: 04/01/2013] [Indexed: 12/31/2022]
Abstract
The dynamic, posttranslational modification of proteins with a small ubiquitin-like modifier (SUMO) tag has been recognized as an important cellular regulatory mechanism relevant to a number of cancers as well as normal embryonic development. As part of a program aimed toward the identification of inhibitors of SUMO-conjugating enzymes, we developed a microfluidic electrophoretic mobility shift assay to monitor sumoylation events in real time. We disclose herein the use of this assay to identify a cell-permeable compound capable of blocking the transfer of SUMO-1 from the E2 enzyme Ubc9 to the substrate. We screened a small collection of compounds and identified an oxygenated flavonoid derivative that inhibits sumoylation in vitro. Next, we carried out an in-depth mechanistic analysis that ruled out many common false-positive mechanisms such as aggregation or alkylation. Furthermore, we report that this flavonoid inhibits a single step in the sumoylation cascade: the transfer of SUMO from the E2 enzyme (Ubc9) thioester conjugate to the substrate. In addition to having a unique mechanism of action, this inhibitor has a discrete structure-activity relationship uncharacteristic of a promiscuous inhibitor. Cell-based studies showed that the flavonoid inhibits the sumoylation of topoisomerase-I in response to camptothecin treatment in two different breast cancer cell lines, while isomeric analogs are inactive. Importantly, this compound blocks sumoylation while not affecting ubiquitylation in cells. This work identifies a point of entry for pharmacologic inhibition of the sumoylation cascade and may serve as the basis for continued study of additional pharmacophores that modulate SUMO-conjugating enzymes such as Ubc9.
Collapse
Affiliation(s)
- Yeong Sang Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
35
|
Klein CA, Droll D, Clayton C. SUMOylation in Trypanosoma brucei. PeerJ 2013; 1:e180. [PMID: 24133638 PMCID: PMC3796365 DOI: 10.7717/peerj.180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
Small ubiquitin like modifier (SUMO) proteins are involved in many processes in eukaryotes. We here show that Trypanosoma brucei SUMO (Tb927.5.3210) modifies many proteins. The levels of SUMOylation were unaffected by temperature changes but were increased by severe oxidative stress. We obtained evidence that trypanosome homologues of the SUMO conjugating enzyme Ubc9 (Tb927.2.2460) and the SUMO-specific protease SENP (Tb927.9.2220) are involved in SUMOylation and SUMO removal, respectively.
Collapse
Affiliation(s)
- Cornelia Andrea Klein
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance , Heidelberg , Germany
| | | | | |
Collapse
|
36
|
Scavenger receptors mediate the role of SUMO and Ftz-f1 in Drosophila steroidogenesis. PLoS Genet 2013; 9:e1003473. [PMID: 23637637 PMCID: PMC3630131 DOI: 10.1371/journal.pgen.1003473] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 03/07/2013] [Indexed: 12/31/2022] Open
Abstract
SUMOylation participates in ecdysteroid biosynthesis at the onset of metamorphosis in Drosophila melanogaster. Silencing the Drosophila SUMO homologue smt3 in the prothoracic gland leads to reduced lipid content, low ecdysone titers, and a block in the larval–pupal transition. Here we show that the SR-BI family of Scavenger Receptors mediates SUMO functions. Reduced levels of Snmp1 compromise lipid uptake in the prothoracic gland. In addition, overexpression of Snmp1 is able to recover lipid droplet levels in the smt3 knockdown prothoracic gland cells. Snmp1 expression depends on Ftz-f1 (an NR5A-type orphan nuclear receptor), the expression of which, in turn, depends on SUMO. Furthermore, we show by in vitro and in vivo experiments that Ftz-f1 is SUMOylated. RNAi–mediated knockdown of ftz-f1 phenocopies that of smt3 at the larval to pupal transition, thus Ftz-f1 is an interesting candidate to mediate some of the functions of SUMO at the onset of metamorphosis. Additionally, we demonstrate that the role of SUMOylation, Ftz-f1, and the Scavenger Receptors in lipid capture and mobilization is conserved in other steroidogenic tissues such as the follicle cells of the ovary. smt3 knockdown, as well as ftz-f1 or Scavenger knockdown, depleted the lipid content of the follicle cells, which could be rescued by Snmp1 overexpression. Therefore, our data provide new insights into the regulation of metamorphosis via lipid homeostasis, showing that Drosophila Smt3, Ftz-f1, and SR-BIs are part of a general mechanism for uptake of lipids such as cholesterol, required during development in steroidogenic tissues. Steroid hormones are cholesterol derivates that control many aspects of animal physiology, including development of the adult organisms, growth, energy storage, and reproduction. In insects, pulses of the steroid hormone ecdysone precede molting and metamorphosis, the regulation of hormonal synthesis being a crucial step that determines animal viability and size. Reduced levels of the small ubiquitin-like modifier SUMO in the prothoracic gland block the synthesis of ecdysone, as SUMO is needed for cholesterol intake. Here we show that SUMO is required for the expression of Scavenger Receptors (Class B, type I). These membrane receptors are necessary for lipid uptake by the gland. Strikingly, their expression is sufficient to recover lipid content when SUMO is removed. The expression of the Scavenger Receptors depends on Ftz-f1, a nuclear transcription factor homologous to mammalian Steroidogenic factor 1 (SF-1). Interestingly, the expression of Ftz-f1 also depends on SUMO and, in addition, Ftz-f1 is SUMOylated. This modification modulates its capacity to activate the Scavenger Receptor Snmp1. The role of SUMO, Scavenger Receptors, and Ftz-f1 on lipid intake is conserved in other tissues that synthesize steroid hormones, such as the ovaries. These factors are conserved in vertebrates, with mutations underlying human disease, so this mechanism to regulate lipid uptake could have implications for human health.
Collapse
|
37
|
Bielska K, Seliga J, Wieczorek E, Kędracka-Krok S, Niedenthal R, Ożyhar A. Alternative sumoylation sites in the Drosophila nuclear receptor Usp. J Steroid Biochem Mol Biol 2012; 132:227-38. [PMID: 22676916 DOI: 10.1016/j.jsbmb.2012.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/24/2012] [Accepted: 05/28/2012] [Indexed: 01/09/2023]
Abstract
The ultraspiracle protein (Usp), together with an ecdysone receptor (EcR) forms a heterodimeric ecdysteroid receptor complex, which controls metamorphosis in Drosophila melanogaster. Although the ecdysteroid receptor is considered to be a source of elements for ecdysteroid inducible gene switches in mammals, nothing is known about posttranslational modifications of the receptor constituents in mammalian cells. Up until now there has been no study about Usp sumoylation. Using Ubc9 fusion-directed sumoylation system, we identified Usp as a new target of SUMO1 and SUMO3 modification. Mutagenesis studies on the fragments of Usp indicated that sumoylation can occur alternatively on several defined Lys residues, i.e. three (Lys16, Lys20, Lys37) in A/B region, one (Lys424) in E region and one (Lys506) in F region. However, sumoylation of one Lys residue within A/B region prevents modification of other residues in this region. This was also observed for Lys residues in carboxyl-terminal fragment of Usp, i.e. comprising E and F regions. Mass spectrometry analysis of the full-length Usp indicated that the main SUMO attachment site is at Lys20. EcR, the heterodimerization partner of Usp, and muristerone A, the EcR ligand, do not influence sumoylation patterns of Usp. Another heterodimerization partner of Usp - HR38 fused with Ubc9 interacts with Usp in HEK293 cells and allows sumoylation of Usp independent of the direct fusion to Ubc9. Taken together, we propose that sumoylation of DmUsp can be an important factor in modulating its activity by changing molecular interactions.
Collapse
Affiliation(s)
- Katarzyna Bielska
- Department of Biochemistry, Faculty of Chemistry, Wrocław University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | | | | | | | | | | |
Collapse
|
38
|
Kravtsova-Ivantsiv Y, Ciechanover A. Non-canonical ubiquitin-based signals for proteasomal degradation. J Cell Sci 2012; 125:539-48. [PMID: 22389393 DOI: 10.1242/jcs.093567] [Citation(s) in RCA: 162] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulated cellular proteolysis is mediated largely by the ubiquitin-proteasome system (UPS). It is a highly specific process that is time- (e.g. cell cycle), compartment- (e.g. nucleus or endoplasmic reticulum) and substrate quality- (e.g. denatured or misfolded proteins) dependent, and allows fast adaptation to changing conditions. Degradation by the UPS is carried out through two successive steps: the substrate is covalently tagged with ubiquitin and subsequently degraded by the 26S proteasome. The accepted 'canonical' signal for proteasomal recognition is a polyubiquitin chain that is anchored to a lysine residue in the target substrate, and is assembled through isopeptide bonds involving lysine 48 of ubiquitin. However, several 'non-canonical' ubiquitin-based signals for proteasomal targeting have also been identified. These include chains anchored to residues other than internal lysine in the substrates, chains assembled through linking residues other than lysine 48 in ubiquitin, and mixed chains made of both ubiquitin and a ubiquitin-like protein. Furthermore, some proteins can be degraded following modification by a single ubiquitin (monoubiquitylation) or multiple single ubiquitins (multiple monoubiquitylation). Finally, some proteins can be proteasomally degraded without prior ubiquitylation (the process is also often referred to as ubiquitination). In this Commentary, we describe these recent findings and discuss the possible physiological roles of these diverse signals. Furthermore, we discuss the possible impact of this signal diversity on drug development.
Collapse
Affiliation(s)
- Yelena Kravtsova-Ivantsiv
- Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Efron Street, Bat Galim, PO Box 9649, Haifa 31096, Israel.
| | | |
Collapse
|
39
|
Alcaide-Loridan C, Jupin I. Ubiquitin and plant viruses, let's play together! PLANT PHYSIOLOGY 2012; 160:72-82. [PMID: 22802610 PMCID: PMC3440231 DOI: 10.1104/pp.112.201905] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
|
40
|
Altmannová V, Kolesár P, Krejčí L. SUMO Wrestles with Recombination. Biomolecules 2012; 2:350-75. [PMID: 24970142 PMCID: PMC4030836 DOI: 10.3390/biom2030350] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 06/27/2012] [Accepted: 07/13/2012] [Indexed: 01/21/2023] Open
Abstract
DNA double-strand breaks (DSBs) comprise one of the most toxic DNA lesions, as the failure to repair a single DSB has detrimental consequences on the cell. Homologous recombination (HR) constitutes an error-free repair pathway for the repair of DSBs. On the other hand, when uncontrolled, HR can lead to genome rearrangements and needs to be tightly regulated. In recent years, several proteins involved in different steps of HR have been shown to undergo modification by small ubiquitin-like modifier (SUMO) peptide and it has been suggested that deficient sumoylation impairs the progression of HR. This review addresses specific effects of sumoylation on the properties of various HR proteins and describes its importance for the homeostasis of DNA repetitive sequences. The article further illustrates the role of sumoylation in meiotic recombination and the interplay between SUMO and other post-translational modifications.
Collapse
Affiliation(s)
| | - Peter Kolesár
- Department of Biology, Masaryk University, Brno 62500, Czech Republic.
| | - Lumír Krejčí
- Department of Biology, Masaryk University, Brno 62500, Czech Republic.
| |
Collapse
|
41
|
Pelisch F, Pozzi B, Risso G, Muñoz MJ, Srebrow A. DNA damage-induced heterogeneous nuclear ribonucleoprotein K sumoylation regulates p53 transcriptional activation. J Biol Chem 2012; 287:30789-99. [PMID: 22825850 DOI: 10.1074/jbc.m112.390120] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heterogeneous nuclear ribonucleoprotein (hnRNP) K is a nucleocytoplasmic shuttling protein that is a key player in the p53-triggered DNA damage response, acting as a cofactor for p53 in response to DNA damage. hnRNP K is a substrate of the ubiquitin E3 ligase MDM2 and, upon DNA damage, is de-ubiquitylated. In sharp contrast with the role and consequences of the other post-translational modifications, nothing is known about the role of SUMO conjugation to hnRNP K in p53 transcriptional co-activation. In the present work, we show that hnRNP K is modified by SUMO in lysine 422 within its KH3 domain, and sumoylation is regulated by the E3 ligase Pc2/CBX4. Most interestingly, DNA damage stimulates hnRNP K sumoylation through Pc2 E3 activity, and this modification is required for p53 transcriptional activation. Abrogation of hnRNP K sumoylation leads to an aberrant regulation of the p53 target gene p21. Our findings link the DNA damage-induced Pc2 activation to the p53 transcriptional co-activation through hnRNP K sumoylation.
Collapse
Affiliation(s)
- Federico Pelisch
- Instituto de Fisiología, Biología Molecular y Neurociencias-Consejo Nacional de Investigaciones Científicas y Técnicas; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales-Universidad de Buenos Aires, Ciudad Universitaria, Pabellón II, Buenos Aires (C1428EHA), Argentina
| | | | | | | | | |
Collapse
|
42
|
The multifaceted oncoprotein Tax: subcellular localization, posttranslational modifications, and NF-κB activation. Adv Cancer Res 2012; 113:85-120. [PMID: 22429853 DOI: 10.1016/b978-0-12-394280-7.00003-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The human T-cell lymphotropic virus type-I (HTLV-I) is the etiologic agent of adult T-cell leukemia/lymphoma (ATL) and of tropical spastic paraparesis/HTLV-I-associated myelopathy. Constitutive NF-κB activation by the viral oncoprotein Tax plays a crucial role in the induction and maintenance of cellular proliferation, transformation, and inhibition of apoptosis. In an attempt to provide a general view of the molecular mechanisms of constitutive Tax-induced NF-κB activation, we summarize in this review the recent body of literature that supports a major role for Tax posttranslational modifications, chiefly ubiquitination, and SUMOylation, in the NF-κB activity of Tax. These modifications indeed participate in the control of Tax subcellular localization and modulate its protein-protein interaction potential. Tax posttranslational modifications, which highlight the ability of HTLV-I to optimize its limited viral genome size, might represent an attractive target for the design of new therapies for ATL.
Collapse
|
43
|
Kolesar P, Sarangi P, Altmannova V, Zhao X, Krejci L. Dual roles of the SUMO-interacting motif in the regulation of Srs2 sumoylation. Nucleic Acids Res 2012; 40:7831-43. [PMID: 22705796 PMCID: PMC3439891 DOI: 10.1093/nar/gks484] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Srs2 DNA helicase of Saccharomyces cerevisiae affects recombination in multiple ways. Srs2 not only inhibits recombination at stalled replication forks but also promotes the synthesis-dependent strand annealing (SDSA) pathway of recombination. Both functions of Srs2 are regulated by sumoylation--sumoylated PCNA recruits Srs2 to the replication fork to disfavor recombination, and sumoylation of Srs2 can be inhibitory to SDSA in certain backgrounds. To understand Srs2 function, we characterize the mechanism of its sumoylation in vitro and in vivo. Our data show that Srs2 is sumoylated at three lysines, and its sumoylation is facilitated by the Siz SUMO ligases. We also show that Srs2 binds to SUMO via a C-terminal SUMO-interacting motif (SIM). The SIM region is required for Srs2 sumoylation, likely by binding to SUMO-charged Ubc9. Srs2's SIM also cooperates with an adjacent PCNA-specific interaction site in binding to sumoylated PCNA to ensure the specificity of the interaction. These two functions of Srs2's SIM exhibit a competitive relationship: sumoylation of Srs2 decreases the interaction between the SIM and SUMO-PCNA, and the SUMO-PCNA-SIM interaction disfavors Srs2 sumoylation. Our findings suggest a potential mechanism for the equilibrium of sumoylated and PCNA-bound pools of Srs2 in cells.
Collapse
Affiliation(s)
- Peter Kolesar
- Department of Biology, National Centre for Biomolecular Research, Masaryk University, 62500 Brno, Czech Republic
| | | | | | | | | |
Collapse
|
44
|
DeSUMOylation Controls Insulin Exocytosis in Response to Metabolic Signals. Biomolecules 2012; 2:269-81. [PMID: 24970137 PMCID: PMC4030845 DOI: 10.3390/biom2020269] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 05/14/2012] [Accepted: 05/16/2012] [Indexed: 12/25/2022] Open
Abstract
The secretion of insulin by pancreatic islet β-cells plays a pivotal role in glucose homeostasis and diabetes. Recent work suggests an important role for SUMOylation in the control of insulin secretion from β-cells. In this paper we discuss mechanisms whereby (de)SUMOylation may control insulin release by modulating β-cell function at one or more key points; and particularly through the acute and reversible regulation of the exocytotic machinery. Furthermore, we postulate that the SUMO-specific protease SENP1 is an important mediator of insulin exocytosis in response to NADPH, a metabolic secretory signal and major determinant of β-cell redox state. Dialysis of mouse β-cells with NADPH efficiently amplifies β-cell exocytosis even when extracellular glucose is low; an effect that is lost upon knockdown of SENP1. Conversely, over-expression of SENP1 itself augments β-cell exocytosis in a redox-dependent manner. Taken together, we suggest that (de)SUMOylation represents an important mechanism that acutely regulates insulin secretion and that SENP1 can act as an amplifier of insulin exocytosis.
Collapse
|
45
|
Wimmer P, Blanchette P, Schreiner S, Ching W, Groitl P, Berscheminski J, Branton PE, Will H, Dobner T. Cross-talk between phosphorylation and SUMOylation regulates transforming activities of an adenoviral oncoprotein. Oncogene 2012; 32:1626-37. [PMID: 22614022 DOI: 10.1038/onc.2012.187] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Since the discovery of post-translational modification (PTM) by the small ubiquitin-related modifiers (SUMOs), a multitude of proteins have been described to be reversibly modified, resulting in the alteration of several cellular pathways. Interestingly, various pathogens gain access to this modification system, although the molecular mechanisms and functional consequences are barely understood. We show here that the adenoviral oncoprotein E1B-55K is a substrate of the SUMO conjugation system, which is directly linked to its C-terminal phosphorylation. This regulative connection is indispensable for modulation of the tumor suppressor p53/chromatin-remodeling factor Daxx by E1B-55K and, consequently, its oncogenic potential in primary mammalian cells. In virus infection, E1B-55K PTMs are necessary for localization to viral transcription/replication sites. Furthermore, we identify the E2 enzyme Ubc9 as an interaction partner of E1B-55K, providing a possible molecular explanation for SUMO-dependent modulation of cellular target proteins. In conclusion, these results for the first time provide evidence how E1B-55K PTMs are regulated and subsequently facilitate exploitation of the host cell SUMOylation machinery.
Collapse
Affiliation(s)
- P Wimmer
- Department of Molecular Virology, Heinrich-Pette-Institute-Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dai Y, Han K, Zou Z, Yan S, Wang Y, Zhang Z. SUMO-1 of mud crab (Scylla paramamosain) in gametogenesis. Gene 2012; 503:260-8. [PMID: 22579467 DOI: 10.1016/j.gene.2012.04.056] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 03/07/2012] [Accepted: 04/18/2012] [Indexed: 11/30/2022]
Abstract
The small ubiquitin-related modifier-1 (SUMO-1) is a member of a family of ubiquitin-related proteins. SUMO pathway, which is involved in gene expression in eukaryotic posttranslational processing, plays important roles in gene expression, genomic stability and the occurrence of cells, development and other biological processes. Scylla paramamosain is one of the important economic breeding crabs in the southeast coast of China. To date, little is known about the distinct roles of SUMO in crustacean, especially in crabs. In the present study, we report the identification and characterization of mud crab, S. paramamosain SUMO-1 (SpSUMO-1) gene using an approach which combines expressed sequence tag (EST) and rapid amplification cDNA end (RACE). The full length cDNA of SpSUMO-1 gene (GenBank: HM581660) is of 732 bp, including a 282 bp open reading frame which encodes a protein of 93 amino acids. Tissue distribution analysis showed that SpSUMO-1 was expressed more abundantly in the ovary than in other tissues (P<0.01). And the expression profiles of SpSUMO-1 in the different gonad developing stages revealed that the highest expression of SpSUMO-1 occurred at proliferation stage, and then decreased gradually as the ovarian development progressed, while in the testis, the expression level of SpSUMO-1 was relatively stable at different stages of testis development. The distribution of SpSUMO-1 mRNA and its protein was observed in the crab gametogenesis by in situ hybridization and immunocytochemical method respectively. In oogenesis, SpSUMO-1 transcripts presented at the cytoplasm and nucleus of oocytes from proliferation stage to primary vitellogenesis stage, but only appeared in the nucleus of oocytes in secondary and tertiary vitellogenesis stages. Meanwhile, SpSUMO-1 protein was localized in the cytoplasm of oogonia and different developing oocytes. On the other hand, the SpSUMO-1 transcript was detected throughout the spermatogenesis, with the strong positive signals of SpSUMO-1 presented at the nuclei of primary and secondary spermatocytes, spermatids and spermatozoa. Interestingly, the positive signals of acrosomal tubules of spermatozoa were also detected. SpSUMO-1 protein was localized in spermatogonium, primary spermatocyte, secondary spermatocyte and spermatid, but the positive signal was only detected in the nucleus of spermatozoa. All these results suggested that SUMO-1 may play essential roles in the gametogenesis of the crustacea.
Collapse
Affiliation(s)
- Yanbin Dai
- Key Laboratory of Healthy Mariculture in the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen 361021, China
| | | | | | | | | | | |
Collapse
|
47
|
Zhou W, Hannoun Z, Jaffray E, Medine CN, Black JR, Greenhough S, Zhu L, Ross JA, Forbes S, Wilmut I, Iredale JP, Hay RT, Hay DC. SUMOylation of HNF4α regulates protein stability and hepatocyte function. J Cell Sci 2012; 125:3630-5. [PMID: 22505616 PMCID: PMC3445325 DOI: 10.1242/jcs.102889] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The coordination of signalling pathways within the cell is vital for normal human development and post-natal tissue homeostasis. Gene expression and function is therefore tightly controlled at a number of levels. We investigated the role that post-translational modifications play during human hepatocyte differentiation. In particular, we examined the role of the small ubiquitin-like modifier (SUMO) proteins in this process. We used a human embryonic stem cell (hESC)-based model of hepatocyte differentiation to follow changes in protein SUMOylation. Moreover, to confirm the results derived from our cell-based system, we performed in vitro conjugation assays to characterise SUMO modification of a key liver-enriched transcription factor, HNF4α. Our analyses indicate that SUMOylation plays an important role during hepatocellular differentiation and this is mediated, in part, through regulation of the stability of HNF4α in a ubiquitin-dependent manner. Our study provides a better understanding of SUMOylation during human hepatocyte differentiation and maturation. Moreover, we believe the results will stimulate interest in the differentiation and phenotypic regulation of other somatic cell types.
Collapse
Affiliation(s)
- Wenli Zhou
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh Bio Quarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Manning Fox JE, Hajmrle C, Macdonald PE. Novel roles of SUMO in pancreatic β-cells: thinking outside the nucleus. Can J Physiol Pharmacol 2012; 90:765-70. [PMID: 22486776 DOI: 10.1139/y11-134] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The endocrine pancreas is critically important in the regulation of energy metabolism, with defective insulin secretion from pancreatic islet β-cells a major contributing factor to the development of type 2 diabetes. Small ubiquitin-like modifier (SUMO) proteins have been demonstrated to covalently modify a wide range of target proteins, mediating a broad range of cellular processes. While the effects of SUMOylation on β-cell gene transcription have been previously reviewed, recent reports indicate roles for SUMO outside of the nucleus. In this review we shall focus on the reported non-nuclear roles of SUMOylation in the regulation of β-cells, including SUMOylation as a novel signaling pathway in the acute regulation of insulin secretion.
Collapse
Affiliation(s)
- Jocelyn E Manning Fox
- Department of Pharmacology and Alberta Diabetes Institute, Li Ka Shing Centre, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | | | | |
Collapse
|
49
|
Truong K, Lee TD, Chen Y. Small ubiquitin-like modifier (SUMO) modification of E1 Cys domain inhibits E1 Cys domain enzymatic activity. J Biol Chem 2012; 287:15154-63. [PMID: 22403398 DOI: 10.1074/jbc.m112.353789] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although it is well established that ubiquitin-like modifications are tightly regulated, it has been unclear how their E1 activities are controlled. In this study, we found that the SAE2 subunit of the small ubiquitin-like modifier (SUMO) E1 is autoSUMOylated at residue Lys-236, and SUMOylation was catalyzed by Ubc9 at several additional Lys residues surrounding the catalytic Cys-173 of SAE2. AutoSUMOylation of SAE2 did not affect SUMO adenylation or formation of E1·SUMO thioester, but did significantly inhibit the transfer of SUMO from E1 to E2 and overall SUMO conjugations to target proteins due to the altered interaction between E1 and E2. Upon heat shock, SUMOylation of SAE2 was reduced, which corresponded with an increase in global SUMOylation, suggesting that SUMOylation of the Cys domain of SAE2 is a mechanism for "storing" a pool of E1 that can be quickly activated in response to environmental changes. This study is the first to show how E1 activity is controlled by post-translational modifications, and similar regulation likely exists across the homologous E1s of ubiquitin-like modifications.
Collapse
Affiliation(s)
- Khue Truong
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California 91010, USA
| | | | | |
Collapse
|
50
|
Wang J, Wang Y, Lu L. De-SUMOylation of CCCTC binding factor (CTCF) in hypoxic stress-induced human corneal epithelial cells. J Biol Chem 2012; 287:12469-79. [PMID: 22354964 DOI: 10.1074/jbc.m111.286641] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epigenetic factor CCCTC binding factor (CTCF) plays important roles in the genetic control of cell fate. Previous studies found that CTCF is positively and negatively regulated at the transcriptional level by epidermal growth factor (EGF) and ultraviolet (UV) stimulation, respectively. However, it is unknown whether other stresses modify the CTCF protein. Here, we report that regulation of CTCF by de-SUMOylation is dependent upon hypoxic and oxidative stresses. We found that stimulation of human corneal epithelial cells with hypoxic stress suppressed a high molecular mass form of CTCF (150 kDa), but not a lower molecular weight form of CTCF (130 kDa). Further investigation revealed that the hypoxic stress-suppressed 150-kDa CTCF was a small ubiquitin-related modifier (SUMO)ylated form of the protein. Hypoxic stress-induced de-SUMOylation of human CTCF was associated with lysine 74 and 689 residues, but not to the phosphorylation of CTCF. Overexpression of SENP1 induced de-SUMOylation of CTCF. However, knockdown of SENP1 could not rescue hypoxic stress-induced CTCF de-SUMOylation. Overexpression of SUMO1 and SUMO2 increased SUMOylation of CTCF and partially blocked hypoxic stress-induced CTCF de-SUMOylation, suggesting that free cellular SUMO proteins play roles in regulating hypoxia-induced CTCF de-SUMOylation. In addition, hypoxic stress significantly inhibited PAX6 mRNA and protein expressions by suppression of PAX6-P0 promoter activity. The result was further supported by data showing that knockdown of CTCF significantly enhanced expression of PAX6 and abolished hypoxia-induced suppression of PAX6. Thus, we conclude that hypoxic stress induces de-SUMOylation of CTCF to functionally regulate CTCF activity.
Collapse
Affiliation(s)
- Jie Wang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Torrance, California 90502, USA
| | | | | |
Collapse
|