Journal articles on the topic 'SUMO Protease'

To see the other types of publications on this topic, follow the link: SUMO Protease.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'SUMO Protease.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Yang, Wei, Liangli Wang, and Wulf Paschen. "Development of a High-Throughput Screening Assay for Inhibitors of Small Ubiquitin-Like Modifier Proteases." Journal of Biomolecular Screening 18, no. 5 (March 7, 2013): 621–28. http://dx.doi.org/10.1177/1087057113479971.

Full text
Abstract:
Small ubiquitin-like modifier (SUMO1–3) is a small group of proteins that are ligated to lysine residues in target proteins. SUMO conjugation is a highly dynamic process, as SUMOylated proteins are rapidly deconjugated by SUMO proteases. SUMO conjugation/deconjugation plays pivotal roles in major cellular pathways and is associated with a number of pathological conditions. It is therefore of significant clinical interest to develop new strategies to screen for compounds to specifically interfere with SUMO conjugation/deconjugation. Here, we describe a novel high-throughput screening (HTS)–compatible assay to identify inhibitors of SUMO proteases. The assay is based on AlphaScreen technology and uses His-tagged SUMO2 conjugated to Strep-tagged SUMO3 as a SUMO protease substrate. A bacterial SUMOylation system was used to generate this substrate. A three-step purification strategy was employed to yield substrate of high quality. Our data indicated that this unique substrate can be readily detected in the AlphaScreen assays in a dose-dependent manner. Cleavage reactions by SUMO protease with or without inhibitor were monitored based on AlphaScreen signals. Furthermore, the assay was adapted to a 384-well format, and the interplate and interday variability was evaluated in eight 384-well plates. The average Z′ factor was 0.83 ± 0.04, confirming the suitability for HTS applications.
APA, Harvard, Vancouver, ISO, and other styles
2

Mukhopadhyay, Debaditya, Ferhan Ayaydin, Nagamalleswari Kolli, Shyh-Han Tan, Tadashi Anan, Ai Kametaka, Yoshiaki Azuma, Keith D. Wilkinson, and Mary Dasso. "SUSP1 antagonizes formation of highly SUMO2/3-conjugated species." Journal of Cell Biology 174, no. 7 (September 21, 2006): 939–49. http://dx.doi.org/10.1083/jcb.200510103.

Full text
Abstract:
Small ubiquitin-related modifier (SUMO) processing and deconjugation are mediated by sentrin-specific proteases/ubiquitin-like proteases (SENP/Ulps). We show that SUMO-specific protease 1 (SUSP1), a mammalian SENP/Ulp, localizes within the nucleoplasm. SUSP1 depletion within cell lines expressing enhanced green fluorescent protein (EGFP) fusions to individual SUMO paralogues caused redistribution of EGFP-SUMO2 and -SUMO3, particularly into promyelocytic leukemia (PML) bodies. Further analysis suggested that this change resulted primarily from a deficit of SUMO2/3-deconjugation activity. Under these circumstances, PML bodies became enlarged and increased in number. We did not observe a comparable redistribution of EGFP-SUMO1. We have investigated the specificity of SUSP1 using vinyl sulfone inhibitors and model substrates. We found that SUSP1 has a strong paralogue bias toward SUMO2/3 and that it acts preferentially on substrates containing three or more SUMO2/3 moieties. Together, our findings argue that SUSP1 may play a specialized role in dismantling highly conjugated SUMO2 and -3 species that is critical for PML body maintenance.
APA, Harvard, Vancouver, ISO, and other styles
3

Alegre, Kamela O., and David Reverter. "Swapping Small Ubiquitin-like Modifier (SUMO) Isoform Specificity of SUMO Proteases SENP6 and SENP7." Journal of Biological Chemistry 286, no. 41 (August 30, 2011): 36142–51. http://dx.doi.org/10.1074/jbc.m111.268847.

Full text
Abstract:
SUMO proteases can regulate the amounts of SUMO-conjugated proteins in the cell by cleaving off the isopeptidic bond between SUMO and the target protein. Of the six members that constitute the human SENP/ULP protease family, SENP6 and SENP7 are the most divergent members in their conserved catalytic domain. The SENP6 and SENP7 subclass displays a clear proteolytic cleavage preference for SUMO2/3 isoforms. To investigate the structural determinants for such isoform specificity, we have identified a unique sequence insertion in the SENP6 and SENP7 subclass that is essential for their proteolytic activity and that forms a more extensive interface with SUMO during the proteolytic reaction. Furthermore, we have identified a region in the SUMO surface determinant for the SUMO2/3 isoform specificity of SENP6 and SENP7. Double point amino acid mutagenesis on the SUMO surface allows us to swap the specificity of SENP6 and SENP7 between the two SUMO isoforms. Structure-based comparisons combined with biochemical and mutagenesis analysis have revealed Loop 1 insertion in SENP6 and SENP7 as a platform to discriminate between SUMO1 and SUMO2/3 isoforms in this subclass of the SUMO protease family.
APA, Harvard, Vancouver, ISO, and other styles
4

Liu, Yan, Yali Shen, Yang Song, Lei Xu, J. Jefferson P. P. Perry, and Jiayu Liao. "Isopeptidase Kinetics Determination by a Real Time and Sensitive qFRET Approach." Biomolecules 11, no. 5 (April 30, 2021): 673. http://dx.doi.org/10.3390/biom11050673.

Full text
Abstract:
Isopeptidase activity of proteases plays critical roles in physiological and pathological processes in living organisms, such as protein stability in cancers and protein activity in infectious diseases. However, the kinetics of protease isopeptidase activity has not been explored before due to a lack of methodology. Here, we report the development of novel qFRET-based protease assay for characterizing the isopeptidase kinetics of SENP1. The reversible process of SUMOylation in vivo requires an enzymatic cascade that includes E1, E2, and E3 enzymes and Sentrin/SUMO-specific proteases (SENPs), which can act either as endopeptidases that process the pre-SUMO before its conjugation, or as isopeptidases to deconjugate SUMO from its target substrate. We first produced the isopeptidase substrate of CyPet-SUMO1/YPet-RanGAP1c by SUMOylation reaction in the presence of SUMO E1 and E2 enzymes. Then a qFRET analyses of real-time FRET signal reduction of the conjugated substrate of CyPet-SUMO1/YPet-RanGAP1c to free CyPet-SUMO1 and YPet-RanGAP1c by the SENP1 were able to obtain the kinetic parameters, Kcat, KM, and catalytic efficiency (Kcat/KM) of SENP1. This represents a pioneer effort in isopeptidase kinetics determination. Importantly, the general methodology of qFRET-based protease isopeptidase kinetic determination can also be applied to other proteases.
APA, Harvard, Vancouver, ISO, and other styles
5

Lee, Jiwon, Yool Lee, Min Joo Lee, Eonyoung Park, Sung Hwan Kang, Chin Ha Chung, Kun Ho Lee, and Kyungjin Kim. "Dual Modification of BMAL1 by SUMO2/3 and Ubiquitin Promotes Circadian Activation of the CLOCK/BMAL1 Complex." Molecular and Cellular Biology 28, no. 19 (July 21, 2008): 6056–65. http://dx.doi.org/10.1128/mcb.00583-08.

Full text
Abstract:
ABSTRACT Heterodimers of BMAL1 and CLOCK drive rhythmic expression of clock-controlled genes, thereby generating circadian physiology and behavior. Posttranslational modifications of BMAL1 play a key role in modulating the transcriptional activity of the CLOCK/BMAL1 complex during the circadian cycle. Recently, we demonstrated that circadian activation of the heterodimeric transcription factor is accompanied by ubiquitin-dependent proteolysis of BMAL1. Here we show that modification by SUMO localizes BMAL1 exclusively to the promyelocytic leukemia nuclear body (NB) and simultaneously promotes its transactivation and ubiquitin-dependent degradation. Under physiological conditions, BMAL1 was predominantly conjugated to poly-SUMO2/3 rather than SUMO1, and the level of these conjugates underwent rhythmic variation, peaking at times of maximum E-box-mediated circadian transcription. Interestingly, mutation of the sumoylation site (Lys259) of BMAL1 markedly inhibited both its ubiquitination and its proteasome-mediated proteolysis, and these effects were reversed by covalent attachment of SUMO3 to the C terminus of the mutant BMAL1. Consistent with this, SUSP1, a SUMO protease highly specific for SUMO2/3, abolished ubiquitination, as well as sumoylation of BMAL1, while the ubiquitin protease UBP41 blocked BMAL1 ubiquitination but induced accumulation of polysumoylated BMAL1 and its localization to the NB. Furthermore, inhibition of proteasome with MG132 elicited robust nuclear accumulation of SUMO2/3- and ubiquitin-modified BMAL1 that was restricted to the transcriptionally active stage of the circadian cycle. These results indicate that dual modification of BMAL1 by SUMO2/3 and ubiquitin is essential for circadian activation and degradation of the CLOCK/BMAL1 complex.
APA, Harvard, Vancouver, ISO, and other styles
6

Shen, Lin Nan, Changjiang Dong, Huanting Liu, James H. Naismith, and Ronald T. Hay. "The structure of SENP1–SUMO-2 complex suggests a structural basis for discrimination between SUMO paralogues during processing." Biochemical Journal 397, no. 2 (June 28, 2006): 279–88. http://dx.doi.org/10.1042/bj20052030.

Full text
Abstract:
The SUMO (small ubiquitin-like modifier)-specific protease SENP1 (sentrin-specific protease 1) can process the three forms of SUMO to their mature forms and deconjugate SUMO from modified substrates. It has been demonstrated previously that SENP1 processed SUMO-1 more efficiently than SUMO-2, but displayed little difference in its ability to deconjugate the different SUMO paralogues from modified substrates. To determine the basis for this substrate specificity, we have determined the crystal structure of SENP1 in isolation and in a transition-state complex with SUMO-2. The interface between SUMO-2 and SENP1 has a relatively poor complementarity, and most of the recognition is determined by interaction between the conserved C-terminus of SUMO-2 and the cleft in the protease. Although SENP1 is rather similar in structure to the related protease SENP2, these proteases have different SUMO-processing activities. Electrostatic analysis of SENP1 in the region where the C-terminal peptide, removed during maturation, would project indicates that it is the electrostatic complementarity between this region of SENP1 and the C-terminal peptides of the various SUMO paralogues that mediates selectivity.
APA, Harvard, Vancouver, ISO, and other styles
7

Vertegaal, Alfred C. O. "SUMO chains: polymeric signals." Biochemical Society Transactions 38, no. 1 (January 19, 2010): 46–49. http://dx.doi.org/10.1042/bst0380046.

Full text
Abstract:
Ubiquitin and ubiquitin-like proteins are conjugated to a wide variety of target proteins that play roles in all biological processes. Target proteins are conjugated to ubiquitin monomers or to ubiquitin polymers that form via all seven internal lysine residues of ubiquitin. The fate of these target proteins is controlled in a chain architecture-dependent manner. SUMO (small ubiquitin-related modifier) shares the ability of ubiquitin to form chains via internal SUMOylation sites. Interestingly, a SUMO-binding site in Ubc9 is important for SUMO chain synthesis. Similar to ubiquitin–polymer cleavage by USPs (ubiquitin-specific proteases), SUMO chain formation is reversible. SUMO polymers are cleaved by the SUMO proteases SENP6 [SUMO/sentrin/SMT3 (suppressor of mif two 3)-specific peptidase 6], SENP7 and Ulp2 (ubiquitin-like protease 2). SUMO chain-binding proteins including ZIP1, SLX5/8 (synthetic lethal of unknown function 5/8), RNF4 (RING finger protein 4) and CENP-E (centromere-associated protein E) have been identified that interact non-covalently with SUMO chains, thereby regulating target proteins that are conjugated to SUMO multimers. SUMO chains play roles in replication, in the turnover of SUMO targets by the proteasome and during mitosis and meiosis. Thus signalling via polymers is an exciting feature of the SUMO family.
APA, Harvard, Vancouver, ISO, and other styles
8

Xu, Zheng, So Fun Chau, Kwok Ho Lam, Ho Yin Chan, Tzi Bun Ng, and Shannon W. N. Au. "Crystal structure of the SENP1 mutant C603S–SUMO complex reveals the hydrolytic mechanism of SUMO-specific protease." Biochemical Journal 398, no. 3 (August 29, 2006): 345–52. http://dx.doi.org/10.1042/bj20060526.

Full text
Abstract:
SUMO (small ubiquitin-related modifier)-specific proteases catalyse the maturation and de-conjugation processes of the sumoylation pathway and modulate various cellular responses including nuclear metabolism and cell cycle progression. The active-site cysteine residue is conserved among all known SUMO-specific proteases and is not substitutable by serine in the hydrolysis reactions demonstrated previously in yeast. We report here that the catalytic domain of human protease SENP1 (SUMO-specific protease 1) mutant SENP1CC603S carrying a mutation of cysteine to serine at the active site is inactive in maturation and de-conjugation reactions. To further understand the hydrolytic mechanism catalysed by SENP1, we have determined, at 2.8 Å resolution (1 Å=0.1 nm), the X-ray structure of SENP1CC603S–SUMO-1 complex. A comparison of the structure of SENP2–SUMO-1 suggests strongly that SUMO-specific proteases require a self-conformational change prior to cleavage of peptide or isopeptide bond in the maturation and de-conjugation processes respectively. Moreover, analysis of the interface of SENP1 and SUMO-1 has led to the identification of four unique amino acids in SENP1 that facilitate the binding of SUMO-1. By means of an in vitro assay, we further demonstrate a novel function of SENP1 in hydrolysing the thioester linkage in E1-SUMO and E2-SUMO complexes. The results disclose a new mechanism of regulation of the sumoylation pathway by the SUMO-specific proteases.
APA, Harvard, Vancouver, ISO, and other styles
9

Di Bacco, Alessandra, Jian Ouyang, Hsiang-Ying Lee, Andre Catic, Hidde Ploegh, and Grace Gill. "The SUMO-Specific Protease SENP5 Is Required for Cell Division." Molecular and Cellular Biology 26, no. 12 (June 15, 2006): 4489–98. http://dx.doi.org/10.1128/mcb.02301-05.

Full text
Abstract:
ABSTRACT Posttranslational modification of substrates by the small ubiquitin-like modifier, SUMO, regulates diverse biological processes, including transcription, DNA repair, nucleocytoplasmic trafficking, and chromosome segregation. SUMOylation is reversible, and several mammalian homologs of the yeast SUMO-specific protease Ulp1, termed SENPs, have been identified. We demonstrate here that SENP5, a previously uncharacterized Ulp1 homolog, has SUMO C-terminal hydrolase and SUMO isopeptidase activities. In contrast to other SENPs, the C-terminal catalytic domain of SENP5 preferentially processed SUMO-3 compared to SUMO-1 precursors and preferentially removed SUMO-2 and SUMO-3 from SUMO-modified RanGAP1 in vitro. In cotransfection assays, SENP5 preferentially reduced high-molecular-weight conjugates of SUMO-2 compared to SUMO-1 in vivo. Full-length SENP5 localized to the nucleolus. Deletion of the noncatalytic N-terminal domain led to loss of nucleolar localization and increased de-SUMOylation activity in vivo. Knockdown of SENP5 by RNA interference resulted in increased levels of SUMO-1 and SUMO-2/3 conjugates, inhibition of cell proliferation, defects in nuclear morphology, and appearance of binucleate cells, revealing an essential role for SENP5 in mitosis and/or cytokinesis. These findings establish SENP5 as a SUMO-specific protease required for cell division and suggest that mechanisms involving both the catalytic and noncatalytic domains determine the distinct substrate specificities of the mammalian SUMO-specific proteases.
APA, Harvard, Vancouver, ISO, and other styles
10

Dorval, Véronique, Matthew J. Mazzella, Paul M. Mathews, Ronald T. Hay, and Paul E. Fraser. "Modulation of Aβ generation by small ubiquitin-like modifiers does not require conjugation to target proteins." Biochemical Journal 404, no. 2 (May 14, 2007): 309–16. http://dx.doi.org/10.1042/bj20061451.

Full text
Abstract:
The sequential processing of the APP (amyloid precursor protein) by the β- and γ-secretase and generation of the Aβ (amyloid-β) peptide is a primary pathological factor in AD (Alzheimer's disease). Regulation of the processing or turnover of these proteins represents potential targets for the development of AD therapies. Sumoylation is a process by which SUMOs (small ubiquitin-like modifiers) are covalently conjugated to target proteins, resulting in a number of functional consequences. These include regulation of protein–protein interactions, intracellular trafficking and protein stability, which all have the potential to impact on several aspects of the amyloidogenic pathway. The present study examines the effects of overexpression and knockdown of the major SUMO isoforms (SUMO1, 2 and 3) on APP processing and the production of Aβ peptides. SUMO3 overexpression significantly increased Aβ40 and Aβ42 secretion, which was accompanied by an increase in full-length APP and its C-terminal fragments. These effects of SUMO3 were independent of its covalent attachment or chain formation, as mutants lacking the motifs responsible for SUMO chain formation or SUMO conjugation led to similar changes in Aβ. SUMO3 overexpression also up-regulated the expression of the transmembrane protease BACE (β-amyloid-cleaving enzyme), but failed to affect levels of several other unrelated proteins. Suppression of SUMO1 or combined SUMO2+3 by RNA interference did not affect APP levels or Aβ production. These findings confirm a specific effect of SUMO3 overexpression on APP processing and the production of Aβ peptides but also suggest that endogenous sumoylation is not essential and likely plays an indirect role in modulating the amyloid processing pathway.
APA, Harvard, Vancouver, ISO, and other styles
11

Roden, Julie, Leah Eardley, Andrew Hotson, Yajuan Cao, and Mary Beth Mudgett. "Characterization of the Xanthomonas AvrXv4 Effector, a SUMO Protease Translocated into Plant Cells." Molecular Plant-Microbe Interactions® 17, no. 6 (June 2004): 633–43. http://dx.doi.org/10.1094/mpmi.2004.17.6.633.

Full text
Abstract:
Homologs of the Yersinia virulence factor YopJ are found in both animal and plant bacterial pathogens, as well as in plant symbionts. The conservation of this effector family indicates that several pathogens may use YopJ-like proteins to regulate bacteria-host interactions during infection. YopJ and YopJ-like proteins share structural homology with cysteine proteases and are hypothesized to functionally mimic small ubiquitin-like modifier (SUMO) proteases in eukaryotic cells. Strains of the phytopathogenic bacterium Xanthomonas campestris pv. vesicatoria are known to possess four YopJ-like proteins, AvrXv4, AvrBsT, AvrRxv, and XopJ. In this work, we have characterized AvrXv4 to determine if AvrXv4 functions like a SUMO protease in planta during Xanthomonas-plant interactions. We provide evidence that X. campestris pv. vesicatoria secretes and translocates the AvrXv4 protein into plant cells during infection in a type III-dependent manner. Once inside the plant cell, AvrXv4 is localized to the plant cytoplasm. By performing AvrXv4 deletion and mutational analysis, we have identified amino acids required for type III delivery and for host recognition. We show that AvrXv4 recognition by resistant plants requires a functional protease catalytic core, the domain that is conserved in all of the putative YopJ-like cysteine proteases. We also show that AvrXv4 expression in planta leads to a reduction in SUMO-modified proteins, demonstrating that AvrXv4 possesses SUMO isopeptidase activity. Overall, our studies reveal that the YopJ-like effector AvrXv4 encodes a type III SUMO protease effector that is active in the cytoplasmic compartment of plant cells.
APA, Harvard, Vancouver, ISO, and other styles
12

XU, Zheng, and Shannon W. N. AU. "Mapping residues of SUMO precursors essential in differential maturation by SUMO-specific protease, SENP1." Biochemical Journal 386, no. 2 (February 22, 2005): 325–30. http://dx.doi.org/10.1042/bj20041210.

Full text
Abstract:
SUMO (small ubiquitin-related modifier) is a member of the ubiquitin-like protein family that regulates cellular function of a variety of target proteins. SUMO proteins are expressed as their precursor forms. Cleavage of the residues after the ‘GG’ region of these precursors by SUMO-specific proteases in maturation is a prerequisite for subsequent sumoylation. To understand further this proteolytic processing, we expressed and purified SENP1 (sentrin-specific protease 1), one of the SUMO-specific proteases, using an Escherichia coli expression system. We show that SENP1 is capable of processing all SUMO-1, -2 and -3 in vitro; however, the proteolytic efficiency of SUMO-1 is the highest followed by SUMO-2 and -3. We demonstrate further that the catalytic domain of SENP1 (SENP1C) alone can determine the substrate specificity towards SUMO-1, -2 and -3. Replacement of the C-terminal fragments after the ‘GG’ region of SUMO-1 and -2 precursors with that of the SUMO-3, indicates that the C-terminal fragment is essential for efficient maturation. In mutagenesis analysis, we further map two residues immediately after the ‘GG’ region, which determine the differential maturation. Distinct patterns of tissue distribution of SENP1, SUMO-1, -2 and -3 are characterized. Taken together, we suggest that the observed differential maturation process has its physiological significance in the regulation of the sumoylation pathway.
APA, Harvard, Vancouver, ISO, and other styles
13

Peek, Jennifer, Catherine Harvey, Dreux Gray, Danny Rosenberg, Likhitha Kolla, Reuben Levy-Myers, Rui Yin, Jonathan L. McMurry, and Oliver Kerscher. "SUMO targeting of a stress-tolerant Ulp1 SUMO protease." PLOS ONE 13, no. 1 (January 19, 2018): e0191391. http://dx.doi.org/10.1371/journal.pone.0191391.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Elmore, Zachary C., Megan Donaher, Brooke C. Matson, Helen Murphy, Jason W. Westerbeck, and Oliver Kerscher. "Sumo-dependent substrate targeting of the SUMO protease Ulp1." BMC Biology 9, no. 1 (2011): 74. http://dx.doi.org/10.1186/1741-7007-9-74.

Full text
APA, Harvard, Vancouver, ISO, and other styles
15

Kroetz, Mary B., Dan Su, and Mark Hochstrasser. "Essential Role of Nuclear Localization for Yeast Ulp2 SUMO Protease Function." Molecular Biology of the Cell 20, no. 8 (April 15, 2009): 2196–206. http://dx.doi.org/10.1091/mbc.e08-10-1090.

Full text
Abstract:
The SUMO protein is covalently attached to many different substrates throughout the cell. This modification is rapidly reversed by SUMO proteases. The Saccharomyces cerevisiae SUMO protease Ulp2 is a nuclear protein required for chromosome stability and cell cycle restart after checkpoint arrest. Ulp2 is related to the human SENP6 protease, also a nuclear protein. All members of the Ulp2/SENP6 family of SUMO proteases have large but poorly conserved N-terminal domains (NTDs) adjacent to the catalytic domain. Ulp2 also has a long C-terminal domain (CTD). We show that CTD deletion has modest effects on yeast growth, but poly-SUMO conjugates accumulate. In contrast, the NTD is essential for Ulp2 function and is required for nuclear targeting. Two short, widely separated sequences within the NTD confer nuclear localization. Efficient Ulp2 import into the nucleus requires the β-importin Kap95, which functions on classical nuclear-localization signal (NLS)-bearing substrates. Remarkably, replacement of the entire >400-residue NTD by a heterologous NLS results in near-normal Ulp2 function. These data demonstrate that nuclear localization of Ulp2 is crucial in vivo, yet only small segments of the NTD provide the key functional elements, explaining the minimal sequence conservation of the NTDs in the Ulp2/SENP6 family of enzymes.
APA, Harvard, Vancouver, ISO, and other styles
16

Cheng, Jialin, Min Su, Yunfeng Jin, Qinghua Xi, Yan Deng, Jie Chen, Wei Wang, et al. "Upregulation of SENP3/SMT3IP1 promotes epithelial ovarian cancer progression and forecasts poor prognosis." Tumor Biology 39, no. 3 (March 2017): 101042831769454. http://dx.doi.org/10.1177/1010428317694543.

Full text
Abstract:
As a crucial member of the small ubiquitin-like modifier system, SUMO-specific protease 3, was identified to be essential for cell proliferation and ribosomal RNA processing. Recent studies showed that SUMO-specific protease 3 was elevated in ovarian cancer compared to normal tissue samples. However, the connection between SUMO-specific protease 3-specific expression and clinicopathological parameters of epithelial ovarian cancer, as well as the physiologically potential role of SUMO-specific protease 3 in epithelial ovarian cancer remained unclear. In this study, an analysis of 124 paraffin-embedded slices by immunohistochemistry indicated that SUMO-specific protease 3 expression was positively correlated with the International Federation of Gynecology and Obstetrics stages (p = 0.025), tumor grade (p = 0.004), and lymph node metastasis (p = 0.001) and was also a critical prognostic factor for the overall survival of epithelial ovarian cancer patients, as revealed by Kaplan–Meier curve analysis. Knockdown of SUMO-specific protease 3 weakened the proliferation, migration, and invasion capability of ovarian cancer cells, down-regulated the expression of Proliferating Cell Nuclear Antigen, Forkhead Box C2, and N-cadherin, and resulted in upregulation of p21 and E-cadherin. Consistent with our results, SUMO-specific protease 3 had been verified to promote cell proliferation, metastasis, and tumorigenesis in multiple malignant cancers, which was a redox-sensitive molecule mediating the epithelial–mesenchymal transition. Collectively, our findings for the first time specifically supported that SUMO-specific protease 3 might play an important role in the regulation of epithelial ovarian cancer progression and could serve as a potential biomarker for prognosis as well as provide a promising therapeutic target against epithelial ovarian cancer.
APA, Harvard, Vancouver, ISO, and other styles
17

Au, S. W., Z. Xu, K. H. Lam, and C. S. F. Chau. "Differential maturation of SUMO precursors by SUMO-specific protease, SENP1." Acta Crystallographica Section A Foundations of Crystallography 61, a1 (August 23, 2005): c208. http://dx.doi.org/10.1107/s0108767305091142.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

Kang, Heejung, Eui Tae Kim, Hye-Ra Lee, Jung-Jin Park, Yoon Young Go, Cheol Yong Choi, and Jin-Hyun Ahn. "Inhibition of SUMO-independent PML oligomerization by the human cytomegalovirus IE1 protein." Journal of General Virology 87, no. 8 (August 1, 2006): 2181–90. http://dx.doi.org/10.1099/vir.0.81787-0.

Full text
Abstract:
In human cytomegalovirus-infected cells, the immediate-early IE1 protein disrupts the subnuclear structures known as the PML oncogenic domains or PODs, via the induction of PML desumoylation. This activity correlates with the functions of IE1 in transcriptional regulation and in the stimulation of lytic infection. Here, the effects of IE1 in induction of desumoylation of PML were characterized. IE1 did not interfere with the formation of sumoylated forms of PML in vitro. In in vitro assays using the sumoylated proteins, a SUMO-specific protease SENP1 desumoylated both PML and IE1. However, the IE1 proteins generated from bacteria or insect cells were unable to desumoylate PML in the same conditions. Although both IE1 and SUMO proteases such as SENP1, Axam and SuPr-1 efficiently desumoylated PML in co-transfection assays, they exerted different effects on the localization of PML. In cells transfected with either SENP1 or SuPr-1, the number of PML foci was reduced significantly and these remnant PML foci were devoid of SUMO-1 signals. However, in cells co-transfected with both SUMO proteases and IE1, these SUMO-independent PML foci were also completely disrupted. Furthermore, IE1, but not SENP1, was shown to disrupt the PML foci generated via transfection of a sumoylation-deficient mutant of PML. These data suggest that IE1 exhibits neither an inhibitory effect on sumoylation of PML nor intrinsic SUMO protease activity against PML in vitro. The finding that IE1 is capable of disrupting SUMO-independent PML aggregates suggests that inhibition of PML oligomerization by IE1 may play an important role in inducing PML desumoylation in vivo.
APA, Harvard, Vancouver, ISO, and other styles
19

El Mchichi, Bouchra, Tarik Regad, Mohamed-Ali Maroui, Manuel S. Rodriguez, Aleksey Aminev, Sylvie Gerbaud, Nicolas Escriou, Laurent Dianoux, and Mounira K. Chelbi-Alix. "SUMOylation Promotes PML Degradation during Encephalomyocarditis Virus Infection." Journal of Virology 84, no. 22 (September 8, 2010): 11634–45. http://dx.doi.org/10.1128/jvi.01321-10.

Full text
Abstract:
ABSTRACT The promyelocytic leukemia (PML) protein is expressed in the diffuse nuclear fraction of the nucleoplasm and in matrix-associated structures, known as nuclear bodies (NBs). PML NB formation requires the covalent modification of PML to SUMO. The noncovalent interactions of SUMO with PML based on the identification of a SUMO-interacting motif within PML seem to be required for further recruitment within PML NBs of SUMOylated proteins. RNA viruses whose replication takes place in the cytoplasm and is inhibited by PML have developed various strategies to counteract the antiviral defense mediated by PML NBs. We show here that primary fibroblasts derived from PML knockout mice are more sensitive to infection with encephalomyocarditis virus (EMCV), suggesting that the absence of PML results in an increase in EMCV replication. Also, we found that EMCV induces a decrease in PML protein levels both in interferon-treated cells and in PMLIII-expressing cells. Reduction of PML was carried out by the EMCV 3C protease. Indeed, at early times postinfection, EMCV induced PML transfer from the nucleoplasm to the nuclear matrix and PML conjugation to SUMO-1, SUMO-2, and SUMO-3, leading to an increase in PML body size where the viral protease 3C and the proteasome component were found colocalizing with PML within the NBs. This process was followed by PML degradation occurring in a proteasome- and SUMO-dependent manner and did not involve the SUMO-interacting motif of PML. Together, these findings reveal a new mechanism evolved by EMCV to antagonize the PML pathway in the interferon-induced antiviral defense.
APA, Harvard, Vancouver, ISO, and other styles
20

Bailey, Daniel, and Peter O’Hare. "Herpes simplex virus 1 ICP0 co-localizes with a SUMO-specific protease." Journal of General Virology 83, no. 12 (December 1, 2002): 2951–64. http://dx.doi.org/10.1099/0022-1317-83-12-2951.

Full text
Abstract:
Early during infection, the herpes simplex regulatory protein ICP0 promotes the proteasome-dependent degradation of a number of cellular proteins and the loss of a number of SUMO-1-modified protein isoforms, including PML. Recently, ICP0 has been shown to induce the accumulation of conjugated ubiquitin and function as a ubiquitin E3 ligase. However, certain aspects of the biochemistry, cell biology and the links between SUMO-1 conjugation/deconjugation and protein degradation remain unclear. For example, it is not currently known whether SUMO-1 deconjugation is a prerequisite for ubiquitination or degradation and, if so, by what mechanism this may occur. To help address these questions, a SUMO-specific protease (SENP1) was cloned and its expression and localization in relation to ICP0 examined. A cell line was established which constitutively expresses SUMO-1 to facilitate studies of localization and biochemistry. SENP1 localized to the nucleus mainly in discrete subdomains, a subset of which co-localized with the PML bodies. Both ICP0 and SENP1 protease promoted the loss of SUMO-1 from the nucleus, observed both for the endogenous species and the cell line expressing the epitope-tagged SUMO-1. The tagged SUMO-1 was recruited into high molecular mass conjugates in the cell line, and expression of SENP1 promoted loss of these species, including the modified species of PML. Finally, in co-transfection experiments ICP0 promoted the recruitment of SENP1 to nuclear domains, a result which was also observed early during infection. The significance of these findings is discussed in relation to the function of ICP0.
APA, Harvard, Vancouver, ISO, and other styles
21

Schuldt, Alison. "A SUMO protease for stress protection." Nature Reviews Molecular Cell Biology 14, no. 5 (April 18, 2013): 263. http://dx.doi.org/10.1038/nrm3569.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Liu, Linpo, Ying Jiang, Xiaomei Zhang, Xu Wang, Yanbing Wang, Yuzhen Han, George Coupland, et al. "Two SUMO Proteases SUMO PROTEASE RELATED TO FERTILITY1 and 2 Are Required for Fertility in Arabidopsis." Plant Physiology 175, no. 4 (October 24, 2017): 1703–19. http://dx.doi.org/10.1104/pp.17.00021.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Itahana, Yoko, Edward T. H. Yeh, and Yanping Zhang. "Nucleocytoplasmic Shuttling Modulates Activity and Ubiquitination-Dependent Turnover of SUMO-Specific Protease 2." Molecular and Cellular Biology 26, no. 12 (June 15, 2006): 4675–89. http://dx.doi.org/10.1128/mcb.01830-05.

Full text
Abstract:
ABSTRACT Small ubiquitin-related modifier (SUMO) proteins are conjugated to numerous polypeptides in cells, and attachment of SUMO plays important roles in regulating the activity, stability, and subcellular localization of modified proteins. SUMO modification of proteins is a dynamic and reversible process. A family of SUMO-specific proteases catalyzes the deconjugation of SUMO-modified proteins. Members of the Sentrin (also known as SUMO)-specific protease (SENP) family have been characterized with unique subcellular localizations. However, little is known about the functional significance of or the regulatory mechanism derived from the specific localizations of the SENPs. Here we identify a bipartite nuclear localization signal (NLS) and a CRM1-dependent nuclear export signal (NES) in the SUMO protease SENP2. Both the NLS and the NES are located in the nonhomologous domains of SENP2 and are not conserved among other members of the SENP family. Using a series of SENP2 mutants and a heterokaryon assay, we demonstrate that SENP2 shuttles between the nucleus and the cytoplasm and that the shuttling is blocked by mutations in the NES or by treating cells with leptomycin B. We show that SENP2 can be polyubiquitinated in vivo and degraded through proteolysis. Restricting SENP2 in the nucleus by mutations in the NES impairs its polyubiquitination, whereas a cytoplasm-localized SENP2 made by introducing mutations in the NLS can be efficiently polyubiquitinated, suggesting that SENP2 is ubiquitinated in the cytoplasm. Finally, treating cells with MG132 leads to accumulation of polyubiquitinated SENP2, indicating that SENP2 is degraded through the 26S proteolysis pathway. Thus, the function of SENP2 is regulated by both nucleocytoplasmic shuttling and polyubiquitin-mediated degradation.
APA, Harvard, Vancouver, ISO, and other styles
24

Nait Achour, Thiziri, Stéphanie Sentis, Catherine Teyssier, Amandine Philippat, Annick Lucas, Laura Corbo, Vincent Cavaillès, and Stéphan Jalaguier. "Transcriptional Repression of Estrogen Receptor α Signaling by SENP2 in Breast Cancer Cells." Molecular Endocrinology 28, no. 2 (February 1, 2014): 183–96. http://dx.doi.org/10.1210/me.2013-1376.

Full text
Abstract:
Abstract Estrogen receptors (ERs) are ligand-activated transcription factors involved in many physiological and pathological processes, including breast cancer. Their activity is fine-tuned by posttranslational modifications, notably sumoylation. In the present study, we investigated the role of the small ubiquitin-related modifier (SUMO) protease, SUMO1/sentrin/suppressor of Mif 2-specific peptidase 2 (SENP2), in the regulation of ERα activity. We first found SENP2 to significantly repress estradiol-induced transcriptional activity in breast cancer cells (MCF7 and T47D). This effect was observed with a reporter plasmid and on endogenous genes such as TFF1 and CTSD, which were shown to recruit SENP2 in chromatin immunoprecipitation experiments. Using glutathione S-transferase pull-down, coimmunoprecipitation and proximity ligation assays, SENP2 was found to interact with ERα and this interaction to be mediated by the amino-terminal region of the protease and the hinge region of the receptor. Interestingly, we demonstrated that ERα repression by SENP2 is independent of its SUMO protease activity and requires a transcriptional repressive domain located in the amino-terminal end of the protease. Using small interfering RNA assays, we evidenced that this domain recruits the histone deacetylase 3 (HDAC3), to be fully active. Furthermore, using both overexpression and knockdown strategies, we showed that SENP2 robustly represses estrogen-dependent and independent proliferation of MCF7 cells. We provided evidence that this effect requires both the proteolytic and transcriptional activities of SENP2. Altogether, our study unravels a new property for a SUMO protease and identifies SENP2 as a classical transcription coregulator.
APA, Harvard, Vancouver, ISO, and other styles
25

Hattersley, Neil, Linnan Shen, Ellis G. Jaffray, and Ronald T. Hay. "The SUMO protease SENP6 is a direct regulator of PML nuclear bodies." Molecular Biology of the Cell 22, no. 1 (January 2011): 78–90. http://dx.doi.org/10.1091/mbc.e10-06-0504.

Full text
Abstract:
Promyelocytic leukemia protein (PML) is the core component of PML-nuclear bodies (PML NBs). The small ubiquitin-like modifier (SUMO) system (and, in particular, SUMOylation of PML) is a critical component in the formation and regulation of PML NBs. SUMO protease SENP6 has been shown previously to be specific for SUMO-2/3–modified substrates and shows preference for SUMO polymers. Here, we further investigate the substrate specificity of SENP6 and show that it is also capable of cleaving mixed chains of SUMO-1 and SUMO-2/3. Depletion of SENP6 results in accumulation of endogenous SUMO-2/3 and SUMO-1 conjugates, and immunofluorescence analysis shows accumulation of SUMO and PML in an increased number of PML NBs. Although SENP6 depletion drastically increases the size of PML NBs, the organizational structure of the body is not affected. Mutation of the catalytic cysteine of SENP6 results in its accumulation in PML NBs, and biochemical analysis indicates that SUMO-modified PML is a substrate of SENP6.
APA, Harvard, Vancouver, ISO, and other styles
26

Liu, Yan, Yali Shen, Shasha Zheng, and Jiayu Liao. "A novel robust quantitative Förster resonance energy transfer assay for protease SENP2 kinetics determination against its all natural substrates." Molecular BioSystems 11, no. 12 (2015): 3407–14. http://dx.doi.org/10.1039/c5mb00568j.

Full text
Abstract:
SUMOylation (the process of adding the SUMO [small ubiquitin-like modifier] to substrates) is an important post-translational modification of critical proteins in multiple processes. The kinetics parameters of pre-SUMO1-3 by its protease SENP2 is determined by a quantitative FRET assay in real time.
APA, Harvard, Vancouver, ISO, and other styles
27

Ambaye, Nigus D. "Noncovalent structure of SENP1 in complex with SUMO2." Acta Crystallographica Section F Structural Biology Communications 75, no. 5 (April 24, 2019): 332–39. http://dx.doi.org/10.1107/s2053230x19004266.

Full text
Abstract:
SUMOylation is a post-translational modification in which a small ubiquitin-like molecule (SUMO) is appended to substrate proteins and is known to influence myriads of biological processes. A delicate interplay between several families of SUMOylation proteins and their substrates ensures the proper level of SUMOylation required for normal cell function. Among the SUMO proteins, SUMO2 is known to form mono-SUMOylated proteins and engage in poly-SUMO chain formation, while sentrin-specific protease 1 (SENP1) is a key enzyme in regulating both events. Determination of the SENP1–SUMO2 interaction is therefore necessary to better understand SUMOylation. In this regard, the current paper reports the noncovalent structure of SENP1 in complex with SUMO2, which was refined to a resolution of 2.62 Å withRandRfreevalues of 22.92% and 27.66%, respectively. The structure shows that SENP1–SUMO2 complex formation is driven largely by polar interactions and limited hydrophobic contacts. The essential C-terminal motif (QQTGG) of SUMO2 is stabilized by a number of specific bonding interactions that enable it to protrude into the catalytic triad of SENP1 and provide the arrangement necessary for maturation of SUMO and deSUMOylation activity. Overall, the structure shows a number of structural details that pinpoint the basis of SENP1–SUMO2 complex formation.
APA, Harvard, Vancouver, ISO, and other styles
28

Verma, Vivek, Anjil K. Srivastava, Catherine Gough, Alberto Campanaro, Moumita Srivastava, Rebecca Morrell, Joshua Joyce, et al. "SUMO enables substrate selectivity by mitogen-activated protein kinases to regulate immunity in plants." Proceedings of the National Academy of Sciences 118, no. 10 (March 1, 2021): e2021351118. http://dx.doi.org/10.1073/pnas.2021351118.

Full text
Abstract:
The versatility of mitogen-activated protein kinases (MAPKs) in translating exogenous and endogenous stimuli into appropriate cellular responses depends on its substrate specificity. In animals, several mechanisms have been proposed about how MAPKs maintain specificity to regulate distinct functional pathways. However, little is known of mechanisms that enable substrate selectivity in plant MAPKs. Small ubiquitin-like modifier (SUMO), a posttranslational modification system, plays an important role in plant development and defense by rapid reprogramming of cellular events. In this study we identified a functional SUMO interaction motif (SIM) in Arabidopsis MPK3 and MPK6 that reveals a mechanism for selective interaction of MPK3/6 with SUMO-conjugated WRKY33, during defense. We show that WRKY33 is rapidly SUMOylated in response to Botrytis cinerea infection and flg22 elicitor treatment. SUMOylation mediates WRKY33 phosphorylation by MPKs and consequent transcription factor activity. Disruption of either WRKY33 SUMO or MPK3/6 SIM sites attenuates their interaction and inactivates WRKY33-mediated defense. However, MPK3/6 SIM mutants show normal interaction with a non-SUMOylated form of another transcription factor, SPEECHLESS, unraveling a role for SUMOylation in differential substrate selectivity by MPKs. We reveal that the SUMO proteases, SUMO PROTEASE RELATED TO FERTILITY1 (SPF1) and SPF2 control WRKY33 SUMOylation and demonstrate a role for these SUMO proteases in defense. Our data reveal a mechanism by which MPK3/6 prioritize molecular pathways by differentially selecting substrates using the SUMO–SIM module during defense responses.
APA, Harvard, Vancouver, ISO, and other styles
29

Li, Shyr-Jiann, and Mark Hochstrasser. "The Ulp1 SUMO isopeptidase." Journal of Cell Biology 160, no. 7 (March 24, 2003): 1069–82. http://dx.doi.org/10.1083/jcb.200212052.

Full text
Abstract:
Protein modification by the ubiquitin-like SUMO protein contributes to many cellular regulatory mechanisms. In Saccharomyces cerevisiae, both sumoylating and desumoylating activities are essential for viability. Of its two known desumoylating enzymes, Ubl-specific protease (Ulp)1 and Ulp2/Smt4, Ulp1 is specifically required for cell cycle progression. A ∼200-residue segment, the Ulp domain (UD), is conserved among Ulps and includes a core cysteine protease domain that is even more widespread. Here we demonstrate that the Ulp1 UD by itself can support wild-type growth rates and in vitro can cleave SUMO from substrates. However, in cells expressing only the UD of Ulp1, many SUMO conjugates accumulate to high levels, indicating that the nonessential Ulp1 NH2-terminal domain is important for activity against a substantial fraction of sumoylated targets. The NH2-terminal domain also includes sequences necessary and sufficient to concentrate Ulp1 at nuclear envelope sites. Remarkably, NH2-terminally deleted Ulp1 variants are able, unlike full-length Ulp1, to suppress defects of cells lacking the divergent Ulp2 isopeptidase. Thus, the NH2-terminal regulatory domain of Ulp1 restricts Ulp1 activity toward certain sumoylated proteins while enabling the cleavage of others. These data define key functional elements of Ulp1 and strongly suggest that subcellular localization is a physiologically significant constraint on SUMO isopeptidase specificity.
APA, Harvard, Vancouver, ISO, and other styles
30

Zhang, Faying, Hui Zheng, Yufan Xian, Haoyue Song, Shengchen Wang, Yueli Yun, Li Yi, and Guimin Zhang. "Profiling Substrate Specificity of the SUMO Protease Ulp1 by the YESS–PSSC System to Advance the Conserved Mechanism for Substrate Cleavage." International Journal of Molecular Sciences 23, no. 20 (October 13, 2022): 12188. http://dx.doi.org/10.3390/ijms232012188.

Full text
Abstract:
SUMO modification is a vital post-translational regulation process in eukaryotes, in which the SUMO protease is responsible for the maturation of the SUMO precursor and the deconjugation of the SUMO protein from modified proteins by accurately cleaving behind the C-terminal Gly–Gly motif. To promote the understanding of the high specificity of the SUMO protease against the SUMO protein as well as to clarify whether the conserved Gly–Gly motif is strictly required for the processing of the SUMO precursor, we systematically profiled the specificity of the S. cerevisiae SUMO protease (Ulp1) on Smt3 at the P2–P1↓P1’ (Gly–Gly↓Ala) position using the YESS–PSSC system. Our results demonstrated that Ulp1 was able to cleave Gly–Gly↓ motif-mutated substrates, indicating that the diglycine motif is not strictly required for Ulp1 cleavage. A structural-modeling analysis indicated that it is the special tapered active pocket of Ulp1 conferred the selectivity of small residues at the P1–P2 position of Smt3, such as Gly, Ala, Ser and Cys, and only which can smoothly deliver the scissile bond into the active site for cleavage. Meanwhile, the P1’ position Ala of Smt3 was found to play a vital role in maintaining Ulp1’s precise cleavage after the Gly–Gly motif and replacing Ala with Gly in this position could expand Ulp1 inclusivity against the P1 and P2 position residues of Smt3. All in all, our studies advanced the traditional knowledge of the SUMO protein, which may provide potential directions for the drug discovery of abnormal SUMOylation-related diseases.
APA, Harvard, Vancouver, ISO, and other styles
31

Ji, Mingfei, Zongtao Chai, Jie Chen, Gang Li, Qiang Li, Miao Li, Yelei Ding, Shaoyong Lu, Guanqun Ju, and Jianquan Hou. "Insights into the Allosteric Effect of SENP1 Q597A Mutation on the Hydrolytic Reaction of SUMO1 via an Integrated Computational Study." Molecules 27, no. 13 (June 28, 2022): 4149. http://dx.doi.org/10.3390/molecules27134149.

Full text
Abstract:
Small ubiquitin-related modifier (SUMO)-specific protease 1 (SENP1) is a cysteine protease that catalyzes the cleavage of the C-terminus of SUMO1 for the processing of SUMO precursors and deSUMOylation of target proteins. SENP1 is considered to be a promising target for the treatment of hepatocellular carcinoma (HCC) and prostate cancer. SENP1 Gln597 is located at the unstructured loop connecting the helices α4 to α5. The Q597A mutation of SENP1 allosterically disrupts the hydrolytic reaction of SUMO1 through an unknown mechanism. Here, extensive multiple replicates of microsecond molecular dynamics (MD) simulations, coupled with principal component analysis, dynamic cross-correlation analysis, community network analysis, and binding free energy calculations, were performed to elucidate the detailed mechanism. Our MD simulations showed that the Q597A mutation induced marked dynamic conformational changes in SENP1, especially in the unstructured loop connecting the helices α4 to α5 which the mutation site occupies. Moreover, the Q597A mutation caused conformational changes to catalytic Cys603 and His533 at the active site, which might impair the catalytic activity of SENP1 in processing SUMO1. Moreover, binding free energy calculations revealed that the Q597A mutation had a minor effect on the binding affinity of SUMO1 to SENP1. Together, these results may broaden our understanding of the allosteric modulation of the SENP1−SUMO1 complex.
APA, Harvard, Vancouver, ISO, and other styles
32

Alonso, Annabel, Sonia D'Silva, Maliha Rahman, Pam B. Meluh, Jacob Keeling, Nida Meednu, Harold J. Hoops, and Rita K. Miller. "The yeast homologue of the microtubule-associated protein Lis1 interacts with the sumoylation machinery and a SUMO-targeted ubiquitin ligase." Molecular Biology of the Cell 23, no. 23 (December 2012): 4552–66. http://dx.doi.org/10.1091/mbc.e12-03-0195.

Full text
Abstract:
Microtubules and microtubule-associated proteins are fundamental for multiple cellular processes, including mitosis and intracellular motility, but the factors that control microtubule-associated proteins (MAPs) are poorly understood. Here we show that two MAPs—the CLIP-170 homologue Bik1p and the Lis1 homologue Pac1p—interact with several proteins in the sumoylation pathway. Bik1p and Pac1p interact with Smt3p, the yeast SUMO; Ubc9p, an E2; and Nfi1p, an E3. Bik1p interacts directly with SUMO in vitro, and overexpression of Smt3p and Bik1p results in its in vivo sumoylation. Modified Pac1p is observed when the SUMO protease Ulp1p is inactivated. Both ubiquitin and Smt3p copurify with Pac1p. In contrast to ubiquitination, sumoylation does not directly tag the substrate for degradation. However, SUMO-targeted ubiquitin ligases (STUbLs) can recognize a sumoylated substrate and promote its degradation via ubiquitination and the proteasome. Both Pac1p and Bik1p interact with the STUbL Nis1p-Ris1p and the protease Wss1p. Strains deleted for RIS1 or WSS1 accumulate Pac1p conjugates. This suggests a novel model in which the abundance of these MAPs may be regulated via STUbLs. Pac1p modification is also altered by Kar9p and the dynein regulator She1p. This work has implications for the regulation of dynein's interaction with various cargoes, including its off-loading to the cortex.
APA, Harvard, Vancouver, ISO, and other styles
33

Smith, Matthew, Vinay Bhaskar, Joseph Fernandez, and Albert J. Courey. "DrosophilaUlp1, a Nuclear Pore-associated SUMO Protease, Prevents Accumulation of Cytoplasmic SUMO Conjugates." Journal of Biological Chemistry 279, no. 42 (August 4, 2004): 43805–14. http://dx.doi.org/10.1074/jbc.m404942200.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Malakhov, Michael P., Michael R. Mattern, Oxana A. Malakhova, Mark Drinker, Stephen D. Weeks, and Tauseef R. Butt. "SUMO fusions and SUMO-specific protease for efficient expression and purification of proteins." Journal of Structural and Functional Genomics 5, no. 1/2 (March 2004): 75–86. http://dx.doi.org/10.1023/b:jsfg.0000029237.70316.52.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Bea, Annika, Constanze Kröber-Boncardo, Manpreet Sandhu, Christine Brinker, and Joachim Clos. "The Leishmania donovani SENP Protease Is Required for SUMO Processing but Not for Viability." Genes 11, no. 10 (October 14, 2020): 1198. http://dx.doi.org/10.3390/genes11101198.

Full text
Abstract:
The protozoan parasite Leishmania donovani is part of an early eukaryotic branch and depends on post-transcriptional mechanisms for gene expression regulation. This includes post-transcriptional protein modifications, such as protein phosphorylation. The presence of genes for protein SUMOylation, i.e., the covalent attachment of small ubiquitin-like modifier (SUMO) polypeptides, in the Leishmania genomes prompted us to investigate the importance of the sentrin-specific protease (SENP) and its putative client, SUMO, for the vitality and infectivity of Leishmania donovani. While SENP null mutants are viable with reduced vitality, viable SUMO null mutant lines could not be obtained. SUMO C-terminal processing is disrupted in SENP null mutants, preventing SUMO from covalent attachment to proteins and nuclear translocation. Infectivity in vitro is not affected by the loss of SENP-dependent SUMO processing. We conclude that SENP is required for SUMO processing, but that functions of unprocessed SUMO are critical for Leishmania viability.
APA, Harvard, Vancouver, ISO, and other styles
36

Vera Rodriguez, Arturo, Steffen Frey, and Dirk Görlich. "Engineered SUMO/protease system identifies Pdr6 as a bidirectional nuclear transport receptor." Journal of Cell Biology 218, no. 6 (April 25, 2019): 2006–20. http://dx.doi.org/10.1083/jcb.201812091.

Full text
Abstract:
Cleavage of affinity tags by specific proteases can be exploited for highly selective affinity chromatography. The SUMO/SENP1 system is the most efficient for such application but fails in eukaryotic expression because it cross-reacts with endogenous proteases. Using a novel selection system, we have evolved the SUMOEu/SENP1Eu pair to orthogonality with the yeast and animal enzymes. SUMOEu fusions therefore remain stable in eukaryotic cells. Likewise, overexpressing a SENP1Eu protease is nontoxic in yeast. We have used the SUMOEu system in an affinity-capture-proteolytic-release approach to identify interactors of the yeast importin Pdr6/Kap122. This revealed not only further nuclear import substrates such as Ubc9, but also Pil1, Lsp1, eIF5A, and eEF2 as RanGTP-dependent binders and thus as export cargoes. We confirmed that Pdr6 functions as an exportin in vivo and depletes eIF5A and eEF2 from cell nuclei. Thus, Pdr6 is a bidirectional nuclear transport receptor (i.e., a biportin) that shuttles distinct sets of cargoes in opposite directions.
APA, Harvard, Vancouver, ISO, and other styles
37

Sun, Xiao-Xin, Yingxiao Chen, Yulong Su, Xiaoyan Wang, Krishna Mohan Chauhan, Juan Liang, Colin J. Daniel, Rosalie C. Sears, and Mu-Shui Dai. "SUMO protease SENP1 deSUMOylates and stabilizes c-Myc." Proceedings of the National Academy of Sciences 115, no. 43 (October 10, 2018): 10983–88. http://dx.doi.org/10.1073/pnas.1802932115.

Full text
Abstract:
Posttranslational modifications play a crucial role in the proper control of c-Myc protein stability and activity. c-Myc can be modified by small ubiquitin-like modifier (SUMO). However, how SUMOylation regulates c-Myc stability and activity remains to be elucidated. The deSUMOylation enzyme, SENP1, has recently been shown to have a prooncogenic role in cancer; however, mechanistic understanding of this is limited. Here we show that SENP1 is a c-Myc deSUMOylating enzyme. SENP1 interacts with and deSUMOylates c-Myc in cells and in vitro. Overexpression of wild-type SENP1, but not its catalytically inactive C603S mutant, markedly stabilizes c-Myc and increases its levels and activity. Knockdown of SENP1 reduces c-Myc levels, induces cell cycle arrest, and drastically suppresses cell proliferation. We further show that c-Myc can be comodified by both ubiquitination and SUMOylation. SENP1-mediated deSUMOylation reduces c-Myc polyubiquitination, suggesting that SUMOylation promotes c-Myc degradation through the proteasome system. Interestingly, SENP1-mediated deSUMOylation promotes the accumulation of monoubiquitinated c-Myc and its phosphorylation at serine 62 and threonine 58. SENP1 is frequently overexpressed, correlating with the high expression of c-Myc, in breast cancer tissues. Together, these results reveal that SENP1 is a crucial c-Myc deSUMOylating enzyme that positively regulates c-Myc’s stability and activity.
APA, Harvard, Vancouver, ISO, and other styles
38

Lau, Yue-Ting K., Vladimir Baytshtok, Tessa A. Howard, Brooke M. Fiala, JayLee M. Johnson, Lauren P. Carter, David Baker, Christopher D. Lima, and Christopher D. Bahl. "Discovery and engineering of enhanced SUMO protease enzymes." Journal of Biological Chemistry 293, no. 34 (July 5, 2018): 13224–33. http://dx.doi.org/10.1074/jbc.ra118.004146.

Full text
APA, Harvard, Vancouver, ISO, and other styles
39

Shin, Eun Ju, Hyun Mi Shin, Eori Nam, Won Seog Kim, Ji‐Hoon Kim, Byung‐Ha Oh, and Yungdae Yun. "DeSUMOylating isopeptidase: a second class of SUMO protease." EMBO reports 13, no. 4 (February 28, 2012): 339–46. http://dx.doi.org/10.1038/embor.2012.3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

Mohideen, Firaz, and Christopher D. Lima. "SUMO Takes Control of a Ubiquitin-Specific Protease." Molecular Cell 30, no. 5 (June 2008): 539–40. http://dx.doi.org/10.1016/j.molcel.2008.05.010.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Su, Dan, and Mark Hochstrasser. "A WLM Protein with SUMO-Directed Protease Activity." Molecular and Cellular Biology 30, no. 15 (June 21, 2010): 3734–36. http://dx.doi.org/10.1128/mcb.00673-10.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Vethantham, Vasupradha, Nishta Rao, and James L. Manley. "Sumoylation Modulates the Assembly and Activity of the Pre-mRNA 3′ Processing Complex." Molecular and Cellular Biology 27, no. 24 (October 8, 2007): 8848–58. http://dx.doi.org/10.1128/mcb.01186-07.

Full text
Abstract:
ABSTRACT Eukaryotic pre-mRNA 3′-end formation is catalyzed by a complex set of factors that must be intricately regulated. In this study, we have discovered a novel role for the small ubiquitin-like modifier SUMO in the regulation of mammalian 3′-end processing. We identified symplekin, a factor involved in complex assembly, and CPSF-73, an endonuclease, as SUMO modification substrates. The major sites of sumoylation in symplekin and CPSF-73 were determined and found to be highly conserved across species. A sumoylation-deficient mutant was defective in rescuing cell viability in symplekin small interfering RNA (siRNA)-treated cells, supporting the importance of this modification in symplekin function. We also analyzed the involvement of sumoylation in 3′-end processing by altering the sumoylation status of nuclear extracts. This was done by the addition of a SUMO protease, which we show interacts with both symplekin and CPSF-73, or by siRNA-mediated depletion of ubc9, the SUMO E2-conjugating enzyme. Both treatments resulted in a marked inhibition of processing. The assembly of a functional polyadenylation complex was also impaired by the SUMO protease. Our identification of two key polyadenylation factors as SUMO targets and of the role of SUMO in enhancing the assembly and activity of the 3′-end-processing complex together reveal an important function for SUMO in the processing of mRNA precursors.
APA, Harvard, Vancouver, ISO, and other styles
43

Psakhye, Ivan, and Dana Branzei. "SMC complexes are guarded by the SUMO protease Ulp2 against SUMO-chain-mediated turnover." Cell Reports 36, no. 5 (August 2021): 109485. http://dx.doi.org/10.1016/j.celrep.2021.109485.

Full text
APA, Harvard, Vancouver, ISO, and other styles
44

Nishida, Tamotsu, and Yoshiji Yamada. "SMT3IP1, a nucleolar SUMO-specific protease, deconjugates SUMO-2 from nucleolar and cytoplasmic nucleophosmin." Biochemical and Biophysical Research Communications 374, no. 2 (September 2008): 382–87. http://dx.doi.org/10.1016/j.bbrc.2008.07.047.

Full text
APA, Harvard, Vancouver, ISO, and other styles
45

Xirodimas, Dimitris P., and David P. Lane. "Targeting a nucleolar SUMO protease for degradation: A mechanism by which ARF induces SUMO conjugation." Cell Cycle 7, no. 21 (November 2008): 3287–91. http://dx.doi.org/10.4161/cc.7.21.7232.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Mukhopadhyay, Debaditya, Alexei Arnaoutov, and Mary Dasso. "The SUMO protease SENP6 is essential for inner kinetochore assembly." Journal of Cell Biology 188, no. 5 (March 8, 2010): 681–92. http://dx.doi.org/10.1083/jcb.200909008.

Full text
Abstract:
We have analyzed the mitotic function of SENP6, a small ubiquitin-like modifier (SUMO) protease that disassembles conjugated SUMO-2/3 chains. Cells lacking SENP6 showed defects in spindle assembly and metaphase chromosome congression. Analysis of kinetochore composition in these cells revealed that a subset of proteins became undetectable on inner kinetochores after SENP6 depletion, particularly the CENP-H/I/K complex, whereas other changes in kinetochore composition mimicked defects previously reported to result from CENP-H/I/K depletion. We further found that CENP-I is degraded through the action of RNF4, a ubiquitin ligase which targets polysumoylated proteins for proteasomal degradation, and that SENP6 stabilizes CENP-I by antagonizing RNF4. Together, these findings reveal a novel mechanism whereby the finely balanced activities of SENP6 and RNF4 control vertebrate kinetochore assembly through SUMO-targeted destabilization of inner plate components.
APA, Harvard, Vancouver, ISO, and other styles
47

Cimarosti, Helena, Emi Ashikaga, Nadia Jaafari, Laura Dearden, Philip Rubin, Kevin A. Wilkinson, and Jeremy M. Henley. "Enhanced SUMOylation and SENP-1 Protein Levels following Oxygen and Glucose Deprivation in Neurones." Journal of Cerebral Blood Flow & Metabolism 32, no. 1 (October 12, 2011): 17–22. http://dx.doi.org/10.1038/jcbfm.2011.146.

Full text
Abstract:
Here, we show that oxygen and glucose deprivation (OGD) causes increased small ubiquitin-like modifier (SUMO)-1 and SUMO-2/3 conjugation to substrate proteins in cultured hippocampal neurones. Surprisingly, the SUMO protease SENP-1, which removes SUMO from conjugated proteins, was also increased by OGD, suggesting that the neuronal response to OGD involves a complex interplay between SUMOylation and deSUMOylation. Importantly, decreasing global SUMOylation in cultured hippocampal neurones by overexpression of the catalytic domain of SENP-1 increased neuronal vulnerability to OGD-induced cell death. Taken together, these results suggest a neuroprotective role for neuronal SUMOylation after OGD.
APA, Harvard, Vancouver, ISO, and other styles
48

Best, Jennifer L., Soula Ganiatsas, Sadhana Agarwal, Austin Changou, Paolo Salomoni, Orian Shirihai, Pamela B. Meluh, Pier Paolo Pandolfi, and Leonard I. Zon. "SUMO-1 Protease-1 Regulates Gene Transcription through PML." Molecular Cell 10, no. 4 (October 2002): 843–55. http://dx.doi.org/10.1016/s1097-2765(02)00699-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Chung, Sung Soo, Byung Yong Ahn, Min Kim, Hye Hun Choi, Ho Seon Park, Shinae Kang, Sang Gyu Park, et al. "Control of Adipogenesis by the SUMO-Specific Protease SENP2." Molecular and Cellular Biology 30, no. 9 (March 1, 2010): 2135–46. http://dx.doi.org/10.1128/mcb.00852-09.

Full text
Abstract:
ABSTRACT Here, we demonstrate that SENP2, a desumoylating enzyme, plays a critical role in the control of adipogenesis. SENP2 expression was markedly increased upon the induction of adipocyte differentiation, and this increase was dependent on protein kinase A activation. Remarkably, knockdown of SENP2 led to a dramatic attenuation of adipogenesis with a marked decrease in PPARγ and C/EBPα mRNA levels. Knockdown of SENP2 also caused a marked reduction in the level of C/EBPβ protein but not in that of C/EBPβ mRNA. Interestingly, sumoylation of C/EBPβ dramatically increased its ubiquitination and destabilization, and this increase could be reversed by SENP2. In addition, overexpression of C/EBPβ could overcome the inhibitory effect of SENP2 knockdown on adipogenesis. Furthermore, SENP2 was absolutely required for adipogenesis of preadipocytes implanted into mice. These results establish a critical role for SENP2 in the regulation of adipogenesis by desumoylation and stabilization of C/EBPβ and in turn by promoting the expression of its downstream effectors, such as PPARγ and C/EBPα.
APA, Harvard, Vancouver, ISO, and other styles
50

Hu, Chenxi, and Xiaodong Jiang. "The SUMO-specific protease family regulates cancer cell radiosensitivity." Biomedicine & Pharmacotherapy 109 (January 2019): 66–70. http://dx.doi.org/10.1016/j.biopha.2018.10.071.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography