SENP3

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 360 Experts worldwide ranked by ideXlab platform

Jie Yang - One of the best experts on this subject based on the ideXlab platform.

  • assessment of SENP3 interacting proteins in hepatocytes treated with diethylnitrosamine by bioid assay
    Acta Biochimica et Biophysica Sinica, 2021
    Co-Authors: Fei Chen, Hongyu Yan, Chu Guo, Huiqin Zhu, Xuxu Sun, Jie Yang
    Abstract:

    SUMOylation of proteins regulates cell behaviors and is reversibly removed by small ubiquitin-like modifier (SUMO)-specific proteases (SENPs). The SENP family member SENP3 is involved in SUMO2/3 deconjugation and has been reported to sense cell stress and accumulate in several human cancer cells and macrophages. We previously reported that SENP3-knockout heterozygous mice showed smaller liver, but the pertinent mechanisms of SENP3 and SUMOylated substrates remain unclear. Thus, in this study, we investigated the interacting proteins with SENP3 and the alteration in hepatocytes treated with the xenobiotic diethylnitrosamine (DEN), which is specifically transformed in the liver and induces DNA double-strand breaks. Our data revealed that a certain amount of SENP3 was present in normal, untreated hepatocytes; however, DEN treatment promoted rapid SENP3 accumulation. SENP3 was mainly localized in the nuclei, and its level was significantly increased in the cytoplasm after 2 h of DEN treatment. The application of the recent proximity-dependent biotinylation (BioID) method led to the identification of 310 SENP3-interacting proteins that were involved in not only gene transcription but also RNA splicing, protein folding, and metabolism. Furthermore, after DEN exposure for a short duration, ribosomal proteins as well as proteins associated with mitochondrial ATP synthesis, membrane transport, and bile acid synthesis, rather than DNA repair proteins, were identified. This study provides insights into the diverse regulatory roles of SENP3, and the BioID method seems to be efficient for identifying physiologically relevant insoluble proteins.

  • SENP3 mediated pparγ2 desumoylation in bm mscs potentiates glucocorticoid induced osteoporosis by promoting adipogenesis and weakening osteogenesis
    Frontiers in Cell and Developmental Biology, 2021
    Co-Authors: Yongxing Zhang, Yang Chen, Hangxiang Sun, Wenkan Zhang, Lingling Zhang, Xin Huang, Jie Yang
    Abstract:

    Glucocorticoid-induced osteoporosis (GIOP) is the most common secondary osteoporosis and reduced bone formation was the main pathological change in GIOP. Our previous studies have shown that there was an imbalance between adipogenic and osteogenic differentiation in GIOP BM-MSCs and peroxisome proliferator-activated receptor γ2 (PPARγ2) played a vital role in this disorders. Here, we reported that there was an increase in ROS level and SENP3 expression in Dex-induced osteoporotic BM-MSCs, and enhanced adipogenesis and weakened osteogenesis in osteoporotic BM-MSCs might be caused by upregulated SENP3. Then we found that SENP3 de-SUMOylated PPARγ2 on K107 site to potentiate adipogenesis and weaken osteogenesis. These results may provide new strategy and target in the clinical diagnosis and treatment of GIOP.

  • SENP3 loss promotes m2 macrophage polarization and breast cancer progression
    Molecular Oncology, 2021
    Co-Authors: Ming Xiao, Xuxu Sun, Yimin Lao, Xueqing Sun, Qi Bian, Jie Yang
    Abstract:

    Tumor-associated macrophages (TAM) play a crucial role in promoting cancer progression. Upon cytokine stimulation, TAM preferentially polarize to the anti-inflammatory and pro-tumor M2 subtype. The mechanism underlying such preferential polarization remains elusive. Here, we report that macrophage-specific deletion of the SUMO-specific protease Sentrin/SUMO-specific protease 3 promotes macrophage polarization towards M2 in bone marrow-derived macrophage (BMDM) induced by interleukin 4 (IL-4)/IL-13 and in an ex vivo model (murine Py8119 cell line), as well as in a mouse orthotopic tumor model. Notably, Sentrin/SUMO-specific protease 3 (SENP3) loss in macrophages accelerated breast cancer malignancy in ex vivo and in vivo models. Mechanistically, we identified Akt Serine/threonine kinase 1 (Akt1) as the substrate of SENP3 and found that the enhanced Akt1 SUMOylation upon SENP3 loss resulted in Akt1 hyper-phosphorylation and activation, which facilitates M2 polarization. Analysis of clinical data showed that a lower level of SENP3 in TAM has a strong negative correlation with the level of the M2 marker CD206, as well as with a worse clinical outcome. Thus, increased Akt1 SUMOylation as a result of SENP3 deficiency modulates polarization of macrophages to the M2 subtype within a breast cancer microenvironment, which in turn promotes tumor progression.

  • SENP3 in monocytes macrophages up regulates tissue factor and mediates lipopolysaccharide induced acute lung injury by enhancing jnk phosphorylation
    Journal of Cellular and Molecular Medicine, 2020
    Co-Authors: Xuelian Chen, Jie Yang, Yimin Lao, Yi Chen
    Abstract:

    The mechanisms underlying coagulation abnormalities in sepsis and septic acute lung injury remain unclear. Tissue factor (TF) initiates coagulation; its production can be regulated by reactive oxygen species (ROS); and monocytes/macrophages produce pathological TF during sepsis. The SUMO2/3 protease SENP3 is redox-sensitive, and SENP3 accumulation in lipopolysaccharide (LPS)-activated macrophages is ROS-dependent. To explore whether SENP3 contributes to LPS-activated coagulation, we used mice with SENP3 conditional knockout (cKO) in myeloid cells. In the model of LPS-induced sepsis, SENP3 cKO mice exhibited less severe acute lung injury than SENP3 fl/fl mice. SENP3 cKO mice exhibited decreased TF expression in monocytes and alveolar macrophages, with consequently compromised coagulation in their blood and lungs. In vitro results showed that ROS-induced SENP3 accumulation contributed to LPS-induced TF expression, which was reduced by JNK inhibitor SP600125. Furthermore, mice injected with LPS following SP600125 (75 mg/kg) treatment showed decreased monocytes/macrophages TF production and alleviated coagulation activation, with less severe lung injury and higher survival rates. Collectively, the results suggest that SENP3 mediates LPS-induced coagulation activation by up-regulating monocyte/macrophage TF production in a JNK-dependent manner. This work provides new insights into ROS regulation of LPS-activated coagulation and reveals a link between SUMOylation and coagulation.

  • SENP3 suppresses osteoclastogenesis by de conjugating sumo2 3 from irf8 in bone marrow derived monocytes
    Cell Reports, 2020
    Co-Authors: Yongxing Zhang, Guoying Deng, Jing Yi, Lianfu Deng, Jie Yang, Kai Yang, Qiugen Wang
    Abstract:

    Summary Bone metabolism depends on the balance between osteoclast-driven bone resorption and osteoblast-mediated bone formation. Diseases like osteoporosis are characterized by increased bone destruction due to partially enhanced osteoclastogenesis. Here, we report that the post-translational SUMO modification is critical for regulating osteoclastogenesis. The expression of the SUMO-specific protease SENP3 is downregulated in osteoclast precursors during osteoclast differentiation. Mice with SENP3 deficiency in bone marrow-derived monocytes (BMDMs) exhibit more severe bone loss due to over-activation of osteoclasts after ovariectomy. Deleting SENP3 in BMDMs promotes osteoclast differentiation. Mechanistically, loss of SENP3 increases interferon regulatory factor 8 (IRF8) SUMO3 modification at the K310 amino acid site, which upregulates expression of the nuclear factor of activated T cell c1 (NFATc1) and osteoclastogenesis. In summary, IRF8 de-SUMO modification mediated by SENP3 suppresses osteoclast differentiation and suggests strategies to treat bone loss diseases.

Jinke Cheng - One of the best experts on this subject based on the ideXlab platform.

  • sumo specific protease 1 is critical for myeloid derived suppressor cell development and function
    Cancer Research, 2019
    Co-Authors: Xian Huang, Qiuju Fan, Guoqiang Chen, Yong Zuo, Hongsheng Tan, Baijun Dong, Xiuzhi Wang, Wei Xue, Jinke Cheng
    Abstract:

    Myeloid-derived suppressor cells (MDSC) can suppress immunity and promote tumorigenesis, and their abundance is associated with poor prognosis. In this study, we show that SUMO1/sentrin-specific peptidase 1 (SENP1) regulates the development and function of MDSC. SENP1 deficiency in myeloid cells promoted MDSC expansion in bone marrow, spleen, and other organs. Senp1−/− MDSC showed stronger immunosuppressive activity than Senp1+/+ MDSC; we observed no defects in the differentiation of myeloid precursor cell in Senp1−/− mice. Mechanistically, SENP1-mediated regulation of MDSC was dependent on STAT3 signaling. We identified CD45 as a specific STAT3 phosphatase in MDSC. CD45 was SUMOylated in MDSC and SENP1 could deconjugate SUMOylated CD45. In Senp1−/− MDSC, CD45 was highly SUMOylated, which reduced its phosphatase activity toward STAT3, leading to STAT3-mediated MDSC development and function. These results reveal a suppressive function of SENP1 in modulating MDSC expansion and function via CD45–STAT3 signaling axis. Significance: These findings show that increased SUMOylation of CD45 via loss of SENP1 suppresses CD45-mediated dephosphorylation of STAT3, which promotes MDSC development and function, leading to tumorigenesis.

  • SENP3 maintains the stability and function of regulatory t cells via bach2 desumoylation
    Nature Communications, 2018
    Co-Authors: Yimin Lao, Xiaolu Teng, Yan Zhou, Feixiang Wang, Xinwei Guo, Siyu Deng, Yuzhou Chang, Zhiduo Liu, Lei Chen, Jinke Cheng
    Abstract:

    Regulatory T (Treg) cells are essential for maintaining immune homeostasis and tolerance, but the mechanisms regulating the stability and function of Treg cells have not been fully elucidated. Here we show SUMO-specific protease 3 (SENP3) is a pivotal regulator of Treg cells that functions by controlling the SUMOylation and nuclear localization of BACH2. Treg cell-specific deletion of SENP3 results in T cell activation, autoimmune symptoms and enhanced antitumor T cell responses. SENP3-mediated BACH2 deSUMOylation prevents the nuclear export of BACH2, thereby repressing the genes associated with CD4+ T effector cell differentiation and stabilizing Treg cell-specific gene signatures. Notably, SENP3 accumulation triggered by reactive oxygen species (ROS) is involved in Treg cell-mediated tumor immunosuppression. Our results not only establish the role of SENP3 in the maintenance of Treg cell stability and function via BACH2 deSUMOylation but also clarify the function of SENP3 in the regulation of ROS-induced immune tolerance.

  • mitotic phosphorylation of SENP3 regulates desumoylation of chromosome associated proteins and chromosome stability
    Cancer Research, 2018
    Co-Authors: Bo Wei, Chao Huang, Bin Liu, Yang Wang, Nansong Xia, Qiuju Fan, Guoqiang Chen, Jinke Cheng
    Abstract:

    Progression of mitotic cell cycle and chromosome condensation and segregation are controlled by posttranslational protein modifications such as phosphorylation and SUMOylation. However, how SUMO isopeptidases (SENP) regulate cell mitotic procession is largely unknown. Here, we demonstrate that precise phosphorylation of SENP3 during mitosis suppresses SENP3 deSUMOylation activity towards chromosome-associated proteins, including topoisomerase IIα (TopoIIα). Cyclin B-dependent kinases 1 and protein phosphatase 1α were identified as the kinase and phosphatase in control of mitotic SENP3 phosphorylation, respectively. SENP3 phosphorylation decreased its interaction with TopoIIα, resulting in reduced SENP3 deSUMOylation activity on TopoIIα. Furthermore, we observed mitotic arrest, increased chromosome instability, and promotion of tumorigenesis in cells expressing a nonphosphorylatable SENP3 mutant. These data show that SENP3 phosphorylation plays a crucial role in regulating the SUMOylation of chromosome-associated proteins and chromosome stability in mitosis.Significance: Phosphorylation of SENP3 regulates SUMOylation of chromosome-associated proteins to maintain genomic stability during mitosis. Cancer Res; 78(9); 2171-8. ©2018 AACR.

  • desumoylation of mkk7 kinase by the sumo2 3 protease SENP3 potentiates lipopolysaccharide induced inflammatory signaling in macrophages
    Journal of Biological Chemistry, 2018
    Co-Authors: Yimin Lao, Kai Yang, Jinke Cheng, Edward T H Yeh, Zhaojun Wang, Xueqing Sun, Qiang Zou, Xuemei Tong, Jie Yang
    Abstract:

    Protein SUMOylation has been reported to play a role in innate immune response, but the enzymes, substrates, and consequences of the specific inflammatory signaling events are largely unknown. Reactive oxygen species (ROS) are abundantly produced during macrophage activation and required for Toll-like receptor 4 (TLR4)-mediated inflammatory signaling. Previously, we demonstrated that SENP3 is a redox-sensitive SUMO2/3 protease. To explore any links between reversible SUMOylation and ROS-related inflammatory signaling in macrophage activation, we generated mice with SENP3 conditional knock-out in myeloid cells. In bacterial lipopolysaccharide (LPS)-induced in vitro and in vivo inflammation models, we found that SENP3 deficiency markedly compromises the activation of TLR4 inflammatory signaling and the production of proinflammatory cytokines in macrophages exposed to LPS. Moreover, SENP3 conditional knock-out mice were significantly less susceptible to septic shock. Of note, SENP3 deficiency was associated with impairment in JNK phosphorylation. We found that MKK7, which selectively phosphorylates JNK, is a SENP3 substrate and that SENP3-mediated deSUMOylation of MKK7 may favor its binding to JNK. Importantly, ROS-dependent SENP3 accumulation and MKK7 deSUMOylation rapidly occurred after LPS stimulation. In conclusion, our findings indicate that SENP3 potentiates LPS-induced TLR4 signaling via deSUMOylation of MKK7 leading to enhancement in JNK phosphorylation and the downstream events. Therefore this work provides novel mechanistic insights into redox regulation of innate immune responses.

  • sumoylation and SENP3 regulate stat3 activation in head and neck cancer
    Oncogene, 2016
    Co-Authors: Zhicheng Zhou, Jinke Cheng, Ming Wang, Edward T H Yeh, M Xiao, Y E Chin, Jie Yang
    Abstract:

    Hyperphosphorylation of signal transducer and activator of transcription 3 (STAT3) has been found in various types of human cancers, including head and neck cancer (HNC). Although smoking is critical in the development and progression of HNC, how tobacco components activate STAT3 is unclear. We demonstrated that exposure of HNC cell lines to a tobacco extract induced a rapid Y705 phosphorylation of STAT3 and a rapid increase in the SUMO protease SENP3 that depended on a simultaneous increase in reactive oxygen species. We identified that SUMOylation at the lysine 451 site facilitated STAT3 binding to the phosphatase TC45 through an SUMO-interacting motif of TC45. SENP3 could thus enhance STAT3 phosphorylation by de-conjugating the SUMO2/3 modification of STAT3. Knocking-down of SENP3 greatly impaired basal and induced STAT3 phosphorylation by tobacco extract or interleukin 6. A correlation between SENP3 protein levels and STAT3 Y705 phosphorylation levels in human laryngeal carcinoma specimens was found, which was more significant in the specimens derived from the smoker patients and with poor clinicopathological parameters. Our data identified SUMOylation as a previously undescribed post-translational modification of STAT3 and SENP3 as a critical positive modulator of tobacco- or cytokine-induced STAT3 activation. These findings provide novel insights into the hyperphosphorylation of STAT3 in development of HNC.

Stefan Muller - One of the best experts on this subject based on the ideXlab platform.

  • flightless i governs cell fate by recruiting the sumo isopeptidase SENP3 to distinct hox genes
    Epigenetics & Chromatin, 2017
    Co-Authors: Arnab Nayak, Anja Reck, Christian Morsczeck, Stefan Muller
    Abstract:

    Despite recent studies on the role of ubiquitin-related SUMO modifier in cell fate decisions, our understanding on precise molecular mechanisms of these processes is limited. Previously, we established that the SUMO isopeptidase SENP3 regulates chromatin assembly of the MLL1/2 histone methyltransferase complex at distinct HOX genes, including the osteogenic master regulator DLX3. A comprehensive mechanism that regulates SENP3 transcriptional function was not understood. Here, we identified flightless-I homolog (FLII), a member of the gelsolin family of actin-remodeling proteins, as a novel regulator of SENP3. We demonstrate that FLII is associated with SENP3 and the MLL1/2 complex. We further show that FLII determines SENP3 recruitment and MLL1/2 complex assembly on the DLX3 gene. Consequently, FLII is indispensible for H3K4 methylation and proper loading of active RNA polymerase II at this gene locus. Most importantly, FLII-mediated SENP3 regulation governs osteogenic differentiation of human mesenchymal stem cells. Altogether, these data reveal a crucial functional interconnection of FLII with the sumoylation machinery that converges on epigenetic regulation and cell fate determination.

  • MOESM1 of Flightless-I governs cell fate by recruiting the SUMO isopeptidase SENP3 to distinct HOX genes
    2017
    Co-Authors: Arnab Nayak, Anja Reck, Christian Morsczeck, Stefan Muller
    Abstract:

    Additional file 1: Fig S1. Proteome map of SENP3 derived from SILAC-based mass spectrometry. (a) Schematic representation of SENP3 proteomics. Equal no. of HeLa cells (as mentioned in “Methods”) either unlabeled or metabolically labeled with amino acid isotope (R6K4) was used for IP. Control IP and SENP3 IP were mixed in a 1:1 ratio and loaded in a SDS-PAGE. The whole lane was cut into several small pieces and processed for mass spec (as described in Materials and Methods section). (b) One representative western blot shows the enrichment of endogenous SENP3 in IP lane that was used for MS analysis. (c) The cytoscape map of SENP3 interactome was accomplished after filtering the whole protein group file (generated from MaxQuant analysis) through 4 tier of following selection criteria—(i) normalized H/L SILAC ratio cutoff was set as 2; that is proteins with minimum twofold enrichment compare to IgG control were considered. (ii) PEP score cutoff was set as (0.0001). PEP score is like p value that represents statistical significance of an observed peptide as a true one. Therefore, smaller PEP score is significant. (iii) Minimum three peptides were considered for any proteins and (iv) reproducibility in both the independent experiments. (d) Cytoscape network of SENP3 interactome obtained from two independent SILAC-MS assays. Details of the generation of the cytoscape map are described in appendix figure S1c. (e) A representative MS/MS spectrum of FLII peptide that was generated by MaxQuant Viewer program. Fig S2. SENP3 protein network. (a) The filtered protein candidates (27) generated from SENP3 proteomics were entered into STRING database as input to check for clustering coefficient. Red arrow indicates the bait, SENP3. (b) Information about FLII protein–protein interaction was extracted from STRING database and combined together with SENP3 network from our experiment. Fig S3. FLII–SENP3 interaction is mostly in the nucleus. (a) Related to Fig. 1. HeLa cells were transfected with Flag-SENP3. Two days after transfection, Flag-agarose bead pull down was performed to check the presence of FLII in the western blot. (b) Subcellular fractionation of HeLa cells was performed according to the user protocol (subcellular protein fractionation kit, ThermoFisher Scientific, catalog no. 78840). Endogenous FLII was immunoprecipitated from cytosolic and nuclear fraction. SDS-PAGE of the immunoprecipitate was performed and probed for indicated antibodies. (c) Subcellular localization of endogenous SENP3 and FLII was studied by immunofluorescence using primary antibodies detecting the respective proteins. Fig S4. FLII interaction with RbBP5. (a) Related to Fig. 3. Endogenous FLII was immunoprecipitated from HeLa cells, and blot was probed against indicated antibodies. (b) Related to Fig. 3. Endogenous SENP3 was immunoprecipitated from HeLa cells, and blot was checked for indicated antibodies. (c) Related to Fig. 3. Endogenous WDR5 was immunoprecipitated, and blot was probed against indicated antibodies. (d) Same as Fig. 1d except RbBP5 construct was used for in vitro transcription/translation. (e) Same as additional file 1, Fig S3a, except Flag-tagged FLII constructs were used to check the interaction of various FLII domains (as indicated in the figure) with RbBP5. Fig S5. SENP3 is not involved in ERα and FLII does not influence SENP3 catalytic activity. (a) Post-transfection (control, SENP3 and FLII siRNA), MCF7 cells were cultured for 3 days in phenol red-free DMEM medium supplemented with 5% charcoal-dextran-stripped fetal bovine serum. Cells were then treated overnight with 100 nM estradiol (E2) before RNA extraction. Data represent the average of triplicates from two biological experiments ± SEM. (b) 72 h after siRNA treatment directed against FLII, cell lysate was prepared in the presence or absence of NEM in the lysis buffer. Equal amount of protein (200μg) of protein from control and FLII siRNA cell lysate was mixed with SUMO2-VS [69] substrate at room temperature for 10 min. The SDS-PAGE was probed by using SENP3 and a loading control tubulin antibody. The asterisk mark represents slow-migrating catalytic active SENP3 form appeared as a result of conjugation between the substrate and SENP3. Right panel shows FLII knockdown efficiency. (c) Same as in (a), except anti-HA antibody (that detects SUMO2-VS substrate) was used. Fig S6. FLII influences MLL1/2 complex assembly on DLX3 gene. (a) Same as Fig. 4b except 5μg of flag-FLII plasmid was transfected to HeLa cells. 48 h. post-transfection cells were fixed and processed for ChIP using rabbit flag antibody. Data represent average of at least two biological experiments performed in duplicate. (b) Related to Fig. 4B. HeLa cell lysate was probed with indicated antibodies to monitor the depletion of FLII and SENP3. (C) Same as Fig. 4e, but DLX3.2 primer was used in qPCR (n = 3, T test, *p 

  • mtor signaling regulates nucleolar targeting of the sumo specific isopeptidase SENP3
    Molecular and Cellular Biology, 2014
    Co-Authors: Nithya Raman, Arnab Nayak, Stefan Muller
    Abstract:

    Ribosome biogenesis is a multistep cellular pathway that involves more than 200 regulatory components to ultimately generate translation-competent 80S ribosomes. The initial steps of this process, particularly rRNA processing, take place in the nucleolus, while later stages occur in the nucleoplasm and cytoplasm. One critical factor of 28S rRNA maturation is the SUMO-isopeptidase SENP3. SENP3 tightly interacts with the nucleolar scaffold protein NPM1 and is associated with nucleolar 60S preribosomes. A central question is how changes in energy supply feed into the regulation of ribosome maturation. Here, we show that the nutrient-sensing mTOR kinase pathway controls the nucleolar targeting of SENP3 by regulating its interaction with NPM1. We define an N-terminal domain in SENP3 as the critical NPM1 binding region and provide evidence that mTOR-mediated phosphorylation of serine/threonine residues within this region fosters the interaction of SENP3 with NPM1. The inhibition of mTOR triggers the nucleolar release of SENP3, thereby likely compromising its activity in rRNA processing. Since mTOR activity is tightly coupled to nutrient availability, we propose that this pathway contributes to the adaptation of ribosome maturation in response to the cellular energy status.

  • the sumo specific isopeptidase SENP3 regulates mll1 mll2 methyltransferase complexes and controls osteogenic differentiation
    Molecular Cell, 2014
    Co-Authors: Arnab Nayak, Christian Morsczeck, Sandra Vialebouroncle, Stefan Muller
    Abstract:

    The ubiquitin-like SUMO system regulates gene expression, but the molecular insights into this process are incomplete. We show that the SUMO-specific isopeptidase SENP3 controls H3K4 methylation by regulating histone-modifying SET1/MLL complexes. SET1/MLL complexes are composed of a histone methyltransferase and the regulatory components WDR5, RbBP5, Ash2L, and DPY-30. MLL1/MLL2 complexes contain menin as additional component and are particularly important for the activation of HOX genes. We demonstrate that SENP3 is associated with MLL1/MLL2 complexes and catalyzes deSUMOylation of RbBP5. This is required for activation of a subset of HOX genes, including the developmental regulator DLX3. In the absence of SENP3, the association of menin and Ash2L with the DLX3 gene is impaired, leading to decreased H3K4 methylation and reduced recruitment of active RNA polymerase II. Importantly, the SENP3-DLX3 pathway dictates osteogenic differentiation of human stem cells, thus delineating the importance of balanced SUMOylation for epigenetic control of gene expression programs.

  • the nucleolar sumo specific protease SENP3 reverses sumo modification of nucleophosmin and is required for rrna processing
    EMBO Reports, 2008
    Co-Authors: Markus Haindl, Thomas Harasim, Dirk Eick, Stefan Muller
    Abstract:

    The ubiquitin-like SUMO system functions by a cyclic process of modification and demodification, and recent data suggest that the nucleolus is a site of sumoylation–desumoylation cycles. For example, the tumour suppressor ARF stimulates sumoylation of nucleolar proteins. Here, we show that the nucleolar SUMO-specific protease SENP3 is associated with nucleophosmin (NPM1), a crucial factor in ribosome biogenesis. SENP3 catalyses desumoylation of NPM1–SUMO2 conjugates in vitro and counteracts ARF-induced modification of NPM1 by SUMO2 in vivo. Intriguingly, depletion of SENP3 by short interfering RNA interferes with nucleolar ribosomal RNA processing and inhibits the conversion of the 32S rRNA species to the 28S form, thus phenocopying the processing defect observed on depletion of NPM1. Moreover, mimicking constitutive modification of NPM1 by SUMO2 interferes with 28S rRNA maturation. These results define SENP3 as an essential factor for ribosome biogenesis and suggest that deconjugation of SUMO2 from NPM1 by SENP3 is critically involved in 28S rRNA maturation.

Yingle Liu - One of the best experts on this subject based on the ideXlab platform.

  • sumo1 sumoylates and SENP3 desumoylates nlrp3 to orchestrate the inflammasome activation
    The FASEB Journal, 2020
    Co-Authors: Luyao Shao, Yan Liu, Wenbiao Wang, Pin Wan, Weiyong Liu, Muhammad Adnan Shereen, Fang Liu, Wen Zhang, Quiping Tan, Yingle Liu
    Abstract:

    The NLRP3 inflammasome regulates innate immune and inflammatory responses by promoting caspase1-dependent induction of pro-inflammatory cytokines. However, aberrant inflammasome activation causes diverse diseases, and thus inflammasome activity must be tightly controlled. Here, we reveal a molecular mechanism underlying the regulation of NLRP3 inflammasome. NLRP3 interacts with SUMO-conjugating enzyme (UBC9), which subsequently promotes small ubiquitin-like modifier 1 (SUMO1) to catalyze NLRP3 SUMOylation at residue Lys204. SUMO1-catalyzed SUMOylation of NLRP3 facilitates ASC oligomerization, inflammasome activation, and interleukin-1β secretion. Moreover, this study also reveals that SUMO-specific protease 3 (SENP3) is required for the deSUMOylation of NLRP3. Interestingly, SENP3 deSUMOylates NLRP3 to attenuate ASC recruitment and speck formation, the NLRP3 inflammasome activation, as well as IL-1β cleavage and secretion. In conclusion, we reveal that SUMO1-catalyzed SUMOylation and SENP3-mediated deSUMOylation of NLRP3 orchestrate the inflammasome activation.

  • sumo1 sumoylates and SENP3 desumoylates nlrp3 to orchestrate the inflammasome activation
    Social Science Research Network, 2018
    Co-Authors: Luyao Shao, Wenbiao Wang, Pin Wan, Weiyong Liu, Muhammad Adnan Shereen, Fang Liu, Wen Zhang, Quiping Tan, Yingle Liu
    Abstract:

    The NLRP3 inflammasome regulates innate immune and inflammatory responses by promoting caspase1-dependent induction of pro-inflammatory cytokines. However, aberrant inflammasome activation causes diverse diseases, and thus inflammasome activity must be tightly controlled. Here, we reveal a distinct mechanism underlying the regulation of NLRP3 inflammasome. NLRP3 interacts with SUMO-conjugating enzyme (UBC9) that subsequently promotes small ubiquitin-like modifier 1 (SUMO1) to catalyze NLRP3 SUMOylation. SUMO1 SUMOylates NLRP3 to promote ASC oligomerization, NLRP inflammasome activation, and interleukin-1β secretion. Additionally, SUMOylation residue K204 is critical for SUMO1-catalyzed NLRP3 SUMOylation and the inflammasome activation. Moreover, SUMO-specific protease 3 (SENP3) is required for the deSUMOylation of NLRP3. Interestingly, SENP3 deSUMOylates NLRP3 to attenuate ASC recruitment and speck formation, the NLRP3 inflammasome activation, and IL-1β cleavage and secretion. In conclusion, we reveal that SUMO1-catalyzed SUMOylation and SENP3-mediated deSUMOylation of NLRP3 orchestrate the inflammasome activation.

Jeremy M. Henley - One of the best experts on this subject based on the ideXlab platform.

  • Ischemia and ischemia-reperfusion in H9C2 cells.
    2019
    Co-Authors: Nadiia Rawlings, Laura Lee, Yasuko Nakamura, Kevin A. Wilkinson, Jeremy M. Henley
    Abstract:

    (A, B) Effects of ischemia on cytosolic SENP3 levels in two separate sets of experiments. SENP3 levels were normalized to β-actin (A) or RhoGDI (B). (C) SENP3 levels in a pooled membrane/nuclear fraction, normalized to VDAC. Raw data were analysed using Students t-test to compare control to ischemia conditions. Data presented as normalized to control values, mean ± SEM. *p

  • Protective role of the deSUMOylating enzyme SENP3 in myocardial ischemia-reperfusion injury
    2019
    Co-Authors: Nadiia Rawlings, Laura Lee, Yasuko Nakamura, Kevin A. Wilkinson, Jeremy M. Henley
    Abstract:

    Interruption of blood supply to the heart is a leading cause of death and disability. However, the molecular events that occur during heart ischemia, and how these changes prime consequent cell death upon reperfusion, are poorly understood. Protein SUMOylation is a post-translational modification that has been strongly implicated in the protection of cells against a variety of stressors, including ischemia-reperfusion. In particular, the SUMO2/3-specific protease SENP3 has emerged as an important determinant of cell survival after ischemic infarct. Here, we used the Langendorff perfusion model to examine changes in the levels and localisation of SUMOylated target proteins and SENP3 in whole heart. We observed a 50% loss of SENP3 from the cytosolic fraction of hearts after preconditioning, a 90% loss after ischemia and an 80% loss after ischemia-reperfusion. To examine these effects further, we performed ischemia and ischemia-reperfusion experiments in the cardiomyocyte H9C2 cell line. Similar to whole hearts, ischemia induced a decrease in cytosolic SENP3. Furthermore, shRNA-mediated knockdown of SENP3 led to an increase in the rate of cell death upon reperfusion. Together, our results indicate that cardiac ischemia dramatically alter levels of SENP3 and suggest that this may a mechanism to promote cell survival after ischemia-reperfusion in heart.

  • increased sumo 2 3 ylation mediated by SENP3 degradation is protective against cadmium induced caspase 3 dependent cytotoxicity
    Journal of Toxicological Sciences, 2017
    Co-Authors: Jia Luo, Laura Lee, Kevin A. Wilkinson, Jeremy M. Henley, Sonam Gurung, Chun Guo
    Abstract:

    Increased post-translational modification of proteins by SUMO-2/3 is a cytoprotective response against cell stress induced by ischaemia and reperfusion. However, it is still unclear what other cell stressors trigger protein SUMOylation, what mechanisms enhance and maintain the enhanced SUMOylation, and if it is a general protective mediator against other cytotoxic stresses. Here, we show increased levels of SUMOylation and decreased levels of the SUMO deconjugating enzyme SENP3 in PC12 cells treated with the toxic heavy metal cadmium. In addition, SENP3 knockdown reduced cadmium-induced caspase 3 cleavage and cell death in PC12 cells, while SENP3 overexpression enhanced cell death. These results suggest that SENP3 is an important regulator of the cellular response to cadmium stress in PC12 cells. Our findings are consistent with previous reports of decreased SENP3 and increased SUMOylation in ischaemia, and imply that the regulation of SENP3 levels and subsequent changes in SUMOylation could be a cytoprotective mechanism against caspase 3-mediated cell death.

  • SENP3 mediated desumoylation of drp1 facilitates interaction with mff to promote cell death
    Scientific Reports, 2017
    Co-Authors: Chun Guo, Kevin A. Wilkinson, Ashley J Evans, Philip Rubin, Jeremy M. Henley
    Abstract:

    The GTPase dynamin-related protein 1 (Drp1) is essential for physiological and pathophysiological mitochondrial fission. DeSUMOylation of Drp1 by the enzyme SENP3 promotes cell death during reperfusion after ischaemia by enhancing Drp1 partitioning to the mitochondrial outer membrane (MOM), which causes cytochrome c release and apoptosis. However, how deSUMOylation recruits Drp1 to the MOM is unknown. Here we show that deSUMOylation selectively promotes Drp1 binding to the MOM resident adaptor protein mitochondrial fission factor (Mff). Consistent with this, preventing Drp1 SUMOylation by mutating the SUMO acceptor sites enhances binding to Mff. Conversely, increasing Drp1 SUMOylation by knocking down SENP3 reduces both Drp1 binding to Mff and stress-induced cytochrome c release. Directly tethering Drp1 to the MOM bypasses the need for Mff to evoke cytochrome c release, and occludes the effect of SENP3 overexpression. Thus, Drp1 deSUMOylation promotes cell death by enhancing Mff-mediated mitochondrial recruitment. These data provide a mechanistic explanation for how the SUMOylation status of Drp1 acts as a key switch in cell death/survival decisions following extreme cell stress.

  • SENP3 mediated desumoylation of dynamin related protein 1 promotes cell death following ischaemia
    The EMBO Journal, 2013
    Co-Authors: Chun Guo, Kevin A. Wilkinson, Jia Luo, Keri L Hildick, Laura Dearden, Jeremy M. Henley
    Abstract:

    Global increases in small ubiquitin-like modifier (SUMO)-2/3 conjugation are a neuroprotective response to severe stress but the mechanisms and specific target proteins that determine cell survival have not been identified. Here, we demonstrate that the SUMO-2/3-specific protease SENP3 is degraded during oxygen/glucose deprivation (OGD), an in vitro model of ischaemia, via a pathway involving the unfolded protein response (UPR) kinase PERK and the lysosomal enzyme cathepsin B. A key target for SENP3-mediated deSUMOylation is the GTPase Drp1, which plays a major role in regulating mitochondrial fission. We show that depletion of SENP3 prolongs Drp1 SUMOylation, which suppresses Drp1-mediated cytochrome c release and caspase-mediated cell death. SENP3 levels recover following reoxygenation after OGD allowing deSUMOylation of Drp1, which facilitates Drp1 localization at mitochondria and promotes fragmentation and cytochrome c release. RNAi knockdown of SENP3 protects cells from reoxygenation-induced cell death via a mechanism that requires Drp1 SUMOylation. Thus, we identify a novel adaptive pathway to extreme cell stress in which dynamic changes in SENP3 stability and regulation of Drp1 SUMOylation are crucial determinants of cell fate.