1
|
Gutierrez-Morton E, Wang Y. The role of SUMOylation in biomolecular condensate dynamics and protein localization. CELL INSIGHT 2024; 3:100199. [PMID: 39399482 PMCID: PMC11467568 DOI: 10.1016/j.cellin.2024.100199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 10/15/2024]
Abstract
As a type of protein post-translational modification, SUMOylation is the process that attaches a small ubiquitin-like modifier (SUMO) to lysine residues of protein substrates. Not only do SUMO and ubiquitin exhibit structure similarity, but the enzymatic cascades for SUMOylation and ubiquitination are also similar. It is well established that protein ubiquitination triggers proteasomal degradation, but the function of SUMOylation remains poorly understood compared to ubiquitination. Recent studies reveal the role of SUMOylation in regulating protein localization, stability, and interaction networks. SUMO can be covalently attached to substrates either as an individual monomer (monoSUMOylation) or as a polymeric SUMO chain (polySUMOylation). Strikingly, mono- and polySUMOylation likely play distinct roles in protein subcellular localization and the assembly/disassembly of biomolecular condensates, which are membraneless cellular compartments with concentrated biomolecules. In this review, we summarize the recent advances in the understanding of the function and regulation of SUMOylation, which could reveal potential therapeutic targets in disease pathogenesis.
Collapse
Affiliation(s)
- Emily Gutierrez-Morton
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306-4300, USA
| | - Yanchang Wang
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306-4300, USA
| |
Collapse
|
2
|
Gao Y, Tan YS, Lin J, Chew LY, Aung HY, Palliyana B, Gujar MR, Lin KY, Kondo S, Wang H. SUMOylation of Warts kinase promotes neural stem cell reactivation. Nat Commun 2024; 15:8557. [PMID: 39419973 PMCID: PMC11487185 DOI: 10.1038/s41467-024-52569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
A delicate balance between neural stem cell (NSC) quiescence and proliferation is important for adult neurogenesis and homeostasis. Small ubiquitin-related modifier (SUMO)-dependent post-translational modifications cause rapid and reversible changes in protein functions. However, the role of the SUMO pathway during NSC reactivation and brain development is not established. Here, we show that the key components of the SUMO pathway play an important role in NSC reactivation and brain development in Drosophila. Depletion of SUMO/Smt3 or SUMO conjugating enzyme Ubc9 results in notable defects in NSC reactivation and brain development, while their overexpression leads to premature NSC reactivation. Smt3 protein levels increase with NSC reactivation, which is promoted by the Ser/Thr kinase Akt. Warts/Lats, the core protein kinase of the Hippo pathway, can undergo SUMO- and Ubc9-dependent SUMOylation at Lys766. This modification attenuates Wts phosphorylation by Hippo, leading to the inhibition of the Hippo pathway, and consequently, initiation of NSC reactivation. Moreover, inhibiting Hippo pathway effectively restores the NSC reactivation defects induced by SUMO pathway inhibition. Overall, our study uncovered an important role for the SUMO-Hippo pathway during Drosophila NSC reactivation and brain development.
Collapse
Affiliation(s)
- Yang Gao
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Ye Sing Tan
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Jiaen Lin
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Liang Yuh Chew
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Htet Yamin Aung
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Brinda Palliyana
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Mahekta R Gujar
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Kun-Yang Lin
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Shu Kondo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku, Katsushika-ku, Tokyo, Japan
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Graduate School - Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Fernandez A, Corvalan K, Santis O, Mendez-Ruette M, Caviedes A, Pizarro M, Gomez MT, Batiz LF, Landgraf P, Kahne T, Rojas-Fernandez A, Wyneken U. Sumoylation in astrocytes induces changes in the proteome of the derived small extracellular vesicles which change protein synthesis and dendrite morphology in target neurons. Brain Res 2024; 1823:148679. [PMID: 37972846 DOI: 10.1016/j.brainres.2023.148679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/01/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Emerging evidence highlights the relevance of the protein post-translational modification by SUMO (Small Ubiquitin-like Modifier) in the central nervous system for modulating cognition and plasticity in health and disease. In these processes, astrocyte-to-neuron crosstalk mediated by extracellular vesicles (EVs) plays a yet poorly understood role. Small EVs (sEVs), including microvesicles and exosomes, contain a molecular cargo of lipids, proteins, and nucleic acids that define their biological effect on target cells. Here, we investigated whether SUMOylation globally impacts the sEV protein cargo. For this, sEVs were isolated from primary cultures of astrocytes by ultracentrifugation or using a commercial sEV isolation kit. SUMO levels were regulated: 1) via plasmids that over-express SUMO, or 2) via experimental conditions that increase SUMOylation, i.e., by using the stress hormone corticosterone, or 3) via the SUMOylation inhibitor 2-D08 (2',3',4'-trihydroxy-flavone, 2-(2,3,4-Trihydroxyphenyl)-4H-1-Benzopyran-4-one). Corticosterone and 2-D08 had opposing effects on the number of sEVs and on their protein cargo. Proteomic analysis showed that increased SUMOylation in corticosterone-treated or plasmid-transfected astrocytes increased the presence of proteins related to cell division, transcription, and protein translation in the derived sEVs. When sEVs derived from corticosterone-treated astrocytes were transferred to neurons to assess their impact on protein synthesis using the fluorescence non-canonical amino acid tagging assay (FUNCAT), we detected an increase in protein synthesis, while sEVs from 2-D08-treated astrocytes had no effect. Our results show that SUMO conjugation plays an important role in the modulation of the proteome of astrocyte-derived sEVs with a potential functional impact on neurons.
Collapse
Affiliation(s)
- Anllely Fernandez
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Katherine Corvalan
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Octavia Santis
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Maxs Mendez-Ruette
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Ariel Caviedes
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Matias Pizarro
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Maria-Teresa Gomez
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Luis Federico Batiz
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile
| | - Peter Landgraf
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Germany, 39120 Magdeburg, Germany
| | - Thilo Kahne
- Institute of Experimental Internal Medicine, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Alejandro Rojas-Fernandez
- Instituto de Medicina & Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Ursula Wyneken
- Centro de Investigación e Innovación Biomédica (CIIB), Facultad de Medicina, Universidad de los Andes, Santiago 7620001, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7620001, Chile.
| |
Collapse
|
4
|
Ninova M, Holmes H, Lomenick B, Fejes Tóth K, Aravin AA. Pervasive SUMOylation of heterochromatin and piRNA pathway proteins. CELL GENOMICS 2023; 3:100329. [PMID: 37492097 PMCID: PMC10363806 DOI: 10.1016/j.xgen.2023.100329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/24/2023] [Accepted: 04/26/2023] [Indexed: 07/27/2023]
Abstract
Genome regulation involves complex protein interactions that are often mediated through post-translational modifications (PTMs). SUMOylation-modification by the small ubiquitin-like modifier (SUMO)-has been implicated in numerous essential processes in eukaryotes. In Drosophila, SUMO is required for viability and fertility, with its depletion from ovaries leading to heterochromatin loss and ectopic transposon and gene activation. Here, we developed a proteomics-based strategy to uncover the Drosophila ovarian "SUMOylome," which revealed that SUMOylation is widespread among proteins involved in heterochromatin regulation and different aspects of the Piwi-interacting small RNA (piRNA) pathway that represses transposons. Furthermore, we show that SUMOylation of several piRNA pathway proteins occurs in a Piwi-dependent manner. Together, these data highlight broad implications of protein SUMOylation in epigenetic regulation and indicate novel roles of this modification in the cellular defense against genomic parasites. Finally, this work provides a resource for the study of SUMOylation in other biological contexts in the Drosophila model.
Collapse
Affiliation(s)
- Maria Ninova
- Department of Biochemistry, University of California Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA 92521, USA
| | - Hannah Holmes
- Department of Biochemistry, University of California Riverside, 3401 Watkins Drive, Boyce Hall, Riverside, CA 92521, USA
| | - Brett Lomenick
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Katalin Fejes Tóth
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| | - Alexei A. Aravin
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Boulevard, Pasadena, CA 91125, USA
| |
Collapse
|
5
|
Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, Zhao X, Hai S, Li S, An Z, Dai L. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (Beijing) 2023; 4:e261. [PMID: 37143582 PMCID: PMC10152985 DOI: 10.1002/mco2.261] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Protein posttranslational modifications (PTMs) refer to the breaking or generation of covalent bonds on the backbones or amino acid side chains of proteins and expand the diversity of proteins, which provides the basis for the emergence of organismal complexity. To date, more than 650 types of protein modifications, such as the most well-known phosphorylation, ubiquitination, glycosylation, methylation, SUMOylation, short-chain and long-chain acylation modifications, redox modifications, and irreversible modifications, have been described, and the inventory is still increasing. By changing the protein conformation, localization, activity, stability, charges, and interactions with other biomolecules, PTMs ultimately alter the phenotypes and biological processes of cells. The homeostasis of protein modifications is important to human health. Abnormal PTMs may cause changes in protein properties and loss of protein functions, which are closely related to the occurrence and development of various diseases. In this review, we systematically introduce the characteristics, regulatory mechanisms, and functions of various PTMs in health and diseases. In addition, the therapeutic prospects in various diseases by targeting PTMs and associated regulatory enzymes are also summarized. This work will deepen the understanding of protein modifications in health and diseases and promote the discovery of diagnostic and prognostic markers and drug targets for diseases.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xina Xiao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yijie Qiu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhiqiang Xu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Chunyu Chen
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Baochen Chong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xinjun Zhao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shan Hai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shuangqing Li
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhenmei An
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Lunzhi Dai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
6
|
Zhu Q, Liang P, Chu C, Zhang A, Zhou W. Protein sumoylation in normal and cancer stem cells. Front Mol Biosci 2022; 9:1095142. [PMID: 36601585 PMCID: PMC9806136 DOI: 10.3389/fmolb.2022.1095142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Stem cells with the capacity of self-renewal and differentiation play pivotal roles in normal tissues and malignant tumors. Whereas stem cells are supposed to be genetically identical to their non-stem cell counterparts, cell stemness is deliberately regulated by a dynamic network of molecular mechanisms. Reversible post-translational protein modifications (PTMs) are rapid and reversible non-genetic processes that regulate essentially all physiological and pathological process. Numerous studies have reported the involvement of post-translational protein modifications in the acquirement and maintenance of cell stemness. Recent studies underscore the importance of protein sumoylation, i.e., the covalent attachment of the small ubiquitin-like modifiers (SUMO), as a critical post-translational protein modification in the stem cell populations in development and tumorigenesis. In this review, we summarize the functions of protein sumoylation in different kinds of normal and cancer stem cells. In addition, we describe the upstream regulators and the downstream effectors of protein sumoylation associated with cell stemness. We also introduce the translational studies aiming at sumoylation to target stem cells for disease treatment. Finally, we propose future directions for sumoylation studies in stem cells.
Collapse
Affiliation(s)
- Qiuhong Zhu
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Panpan Liang
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Cuiying Chu
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Aili Zhang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States,*Correspondence: Aili Zhang, ; Wenchao Zhou,
| | - Wenchao Zhou
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China,*Correspondence: Aili Zhang, ; Wenchao Zhou,
| |
Collapse
|
7
|
Hegde S, Sreejan A, Gadgil CJ, Ratnaparkhi GS. SUMOylation of Dorsal attenuates Toll/NF-κB signaling. Genetics 2022; 221:iyac081. [PMID: 35567478 PMCID: PMC9252280 DOI: 10.1093/genetics/iyac081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022] Open
Abstract
In Drosophila, Toll/NF-κB signaling plays key roles in both animal development and in host defense. The activation, intensity, and kinetics of Toll signaling are regulated by posttranslational modifications such as phosphorylation, SUMOylation, or ubiquitination that target multiple proteins in the Toll/NF-κB cascade. Here, we have generated a CRISPR-Cas9 edited Dorsal (DL) variant that is SUMO conjugation resistant. Intriguingly, embryos laid by dlSCR mothers overcome dl haploinsufficiency and complete the developmental program. This ability appears to be a result of higher transcriptional activation by DLSCR. In contrast, SUMOylation dampens DL transcriptional activation, ultimately conferring robustness to the dorso-ventral program. In the larval immune response, dlSCR animals show an increase in crystal cell numbers, stronger activation of humoral defense genes, and high cactus levels. A mathematical model that evaluates the contribution of the small fraction of SUMOylated DL (1-5%) suggests that it acts to block transcriptional activation, which is driven primarily by DL that is not SUMO conjugated. Our findings define SUMO conjugation as an important regulator of the Toll signaling cascade, in both development and host defense. Our results broadly suggest that SUMO attenuates DL at the level of transcriptional activation. Furthermore, we hypothesize that SUMO conjugation of DL may be part of a Ubc9-dependent mechanism that restrains Toll/NF-κB signaling.
Collapse
Affiliation(s)
- Sushmitha Hegde
- Biology, Indian Institute of Science Education & Research, Pune 411008, India
| | - Ashley Sreejan
- Chemical Engineering and Process Development Division, CSIR—National Chemical Laboratory, Pune 411008, India
| | - Chetan J Gadgil
- Chemical Engineering and Process Development Division, CSIR—National Chemical Laboratory, Pune 411008, India
- CSIR—Institute of Genomics and Integrative Biology, New Delhi 110020, India
| | | |
Collapse
|
8
|
Soory A, Ratnaparkhi GS. SUMOylation of Jun fine-tunes the Drosophila gut immune response. PLoS Pathog 2022; 18:e1010356. [PMID: 35255103 PMCID: PMC8929699 DOI: 10.1371/journal.ppat.1010356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 03/17/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Post-translational modification by the small ubiquitin-like modifier, SUMO can modulate the activity of its conjugated proteins in a plethora of cellular contexts. The effect of SUMO conjugation of proteins during an immune response is poorly understood in Drosophila. We have previously identified that the transcription factor Jra, the Drosophila Jun ortholog and a member of the AP-1 complex is one such SUMO target. Here, we find that Jra is a regulator of the Pseudomonas entomophila induced gut immune gene regulatory network, modulating the expression of a few thousand genes, as measured by quantitative RNA sequencing. Decrease in Jra in gut enterocytes is protective, suggesting that reduction of Jra signaling favors the host over the pathogen. In Jra, lysines 29 and 190 are SUMO conjugation targets, with the JraK29R+K190R double mutant being SUMO conjugation resistant (SCR). Interestingly, a JraSCR fly line, generated by CRISPR/Cas9 based genome editing, is more sensitive to infection, with adults showing a weakened host response and increased proliferation of Pseudomonas. Transcriptome analysis of the guts of JraSCR and JraWT flies suggests that lack of SUMOylation of Jra significantly changes core elements of the immune gene regulatory network, which include antimicrobial agents, secreted ligands, feedback regulators, and transcription factors. Mechanistically, SUMOylation attenuates Jra activity, with the TFs, forkhead, anterior open, activating transcription factor 3 and the master immune regulator Relish being important transcriptional targets. Our study implicates Jra as a major immune regulator, with dynamic SUMO conjugation/deconjugation of Jra modulating the kinetics of the gut immune response. The intestine has a resident population of commensal microorganisms against which the immune machinery is tuned to show low or no reactivity. In contrast, when pathogenic microorganisms are ingested, the gut responds by activating signaling cascades that lead to the killing and clearance of the pathogen. In this study, we examine the role played by the well-known transcription factor Jun in regulating the immune response in the Drosophila gut. We find that loss of Jun leads to the change in intensity and kinetics of the gut immune transcriptome. The transcriptional profile indicates a stronger response when Jun activity is reduced. Also, animals infected with Pseudomonas entomophila live longer when Jun signaling is reduced. Further, we find that Jun is post-translationally modified on Lys29 and Lys190 by SUMO. To understand the effect of SUMO-conjugation of Jun, we create by state-of-the-art CRISPR/Cas9 genome editing a Drosophila line where Jun is resistant to SUMOylation. This line is more sensitive to infection, with a weaker host-defense response. Our data suggest that Jun Signaling favors the pathogen by dampening the immune response. SUMO conjugation of Jun reverses the dampening and strengthens the immune response in favor of the host. Dynamic SUMOylation of Jun thus fine-tunes the gut immune response to pathogens.
Collapse
Affiliation(s)
- Amarendranath Soory
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| | - Girish S. Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| |
Collapse
|
9
|
Sumoylation in Physiology, Pathology and Therapy. Cells 2022; 11:cells11050814. [PMID: 35269436 PMCID: PMC8909597 DOI: 10.3390/cells11050814] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
Sumoylation is an essential post-translational modification that has evolved to regulate intricate networks within emerging complexities of eukaryotic cells. Thousands of target substrates are modified by SUMO peptides, leading to changes in protein function, stability or localization, often by modulating interactions. At the cellular level, sumoylation functions as a key regulator of transcription, nuclear integrity, proliferation, senescence, lineage commitment and stemness. A growing number of prokaryotic and viral proteins are also emerging as prime sumoylation targets, highlighting the role of this modification during infection and in immune processes. Sumoylation also oversees epigenetic processes. Accordingly, at the physiological level, it acts as a crucial regulator of development. Yet, perhaps the most prominent function of sumoylation, from mammals to plants, is its role in orchestrating organismal responses to environmental stresses ranging from hypoxia to nutrient stress. Consequently, a growing list of pathological conditions, including cancer and neurodegeneration, have now been unambiguously associated with either aberrant sumoylation of specific proteins and/or dysregulated global cellular sumoylation. Therapeutic enforcement of sumoylation can also accomplish remarkable clinical responses in various diseases, notably acute promyelocytic leukemia (APL). In this review, we will discuss how this modification is emerging as a novel drug target, highlighting from the perspective of translational medicine, its potential and limitations.
Collapse
|
10
|
Cui LH, Min HJ, Yu SG, Byun MY, Oh TR, Lee A, Yang HW, Kim WT. OsATL38 mediates mono-ubiquitination of the 14-3-3 protein OsGF14d and negatively regulates the cold stress response in rice. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:307-323. [PMID: 34436579 DOI: 10.1093/jxb/erab392] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
One of the major regulatory pathways that permits plants to convert an external stimulus into an internal cellular response within a short period of time is the ubiquitination pathway. In this study, OsATL38 was identified as a low temperature-induced gene that encodes a rice homolog of Arabidopsis Tóxicos en Levadura RING-type E3 ubiquitin (Ub) ligase, which was predominantly localized to the plasma membrane. OsATL38-overexpressing transgenic rice plants exhibited decreased tolerance to cold stress as compared with wild-type rice plants. In contrast, RNAi-mediated OsATL38 knockdown transgenic progeny exhibited markedly increased tolerance to cold stress relative to that of wild-type plants, which indicated a negative role of OsATL38 in response to cold stress. Yeast two-hybrid, in vitro pull-down, and co-immunoprecipitation assays revealed that OsATL38 physically interacted with OsGF14d, a rice 14-3-3 protein. An in vivo target ubiquitination assay indicated that OsGF14d was mono-ubiquitinated by OsATL38. osgf14d knockout mutant plants were more sensitive to cold stress than wild-type rice plants, indicating that OsGF14d is a positive factor in the response to cold stress. These results provide evidence that the RING E3 Ub ligase OsATL38 negatively regulates the cold stress response in rice via mono-ubiquitination of OsGF14d 14-3-3 protein.
Collapse
Affiliation(s)
- Li Hua Cui
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Hye Jo Min
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seong Guan Yu
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Mi Young Byun
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Tae Rin Oh
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Andosung Lee
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Hee Woong Yang
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Woo Taek Kim
- Department of Systems Biology and Division of Life Science, Yonsei University, Seoul 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
11
|
In vitro and in cellulae methods for determining the target protein SUMOylation. Methods Enzymol 2022; 675:397-424. [DOI: 10.1016/bs.mie.2022.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
Chang KC. Influence of Sox protein SUMOylation on neural development and regeneration. Neural Regen Res 2022; 17:477-481. [PMID: 34380874 PMCID: PMC8504373 DOI: 10.4103/1673-5374.320968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
SRY-related HMG-box (Sox) transcription factors are known to regulate central nervous system development and are involved in several neurological diseases. Post-translational modification of Sox proteins is known to alter their functions in the central nervous system. Among the different types of post-translational modification, small ubiquitin-like modifier (SUMO) modification of Sox proteins has been shown to modify their transcriptional activity. Here, we review the mechanisms of three Sox proteins in neuronal development and disease, along with their transcriptional changes under SUMOylation. Across three species, lysine is the conserved residue for SUMOylation. In Drosophila, SUMOylation of SoxN plays a repressive role in transcriptional activity, which impairs central nervous system development. However, deSUMOylation of SoxE and Sox11 plays neuroprotective roles, which promote neural crest precursor formation in Xenopus and retinal ganglion cell differentiation as well as axon regeneration in the rodent. We further discuss a potential translational therapy by SUMO site modification using AAV gene transduction and Clustered regularly interspaced short palindromic repeats-Cas9 technology. Understanding the underlying mechanisms of Sox SUMOylation, especially in the rodent system, may provide a therapeutic strategy to address issues associated with neuronal development and neurodegeneration.
Collapse
Affiliation(s)
- Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Nayak P, Kejriwal A, Ratnaparkhi GS. SUMOylation of Arginyl tRNA Synthetase Modulates the Drosophila Innate Immune Response. Front Cell Dev Biol 2021; 9:695630. [PMID: 34660574 PMCID: PMC8514731 DOI: 10.3389/fcell.2021.695630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
SUMO conjugation of a substrate protein can modify its activity, localization, interaction or function. A large number of SUMO targets in cells have been identified by Proteomics, but biological roles for SUMO conjugation for most targets remains elusive. The multi-aminoacyl tRNA synthetase complex (MARS) is a sensor and regulator of immune signaling. The proteins of this 1.2 MDa complex are targets of SUMO conjugation, in response to infection. Arginyl tRNA Synthetase (RRS), a member of the sub-complex II of MARS, is one such SUMO conjugation target. The sites for SUMO conjugation are Lys 147 and 383. Replacement of these residues by Arg (RRS K147R,K383R ), creates a SUMO conjugation resistant variant (RRS SCR ). Transgenic Drosophila lines for RRS WT and RRS SCR were generated by expressing these variants in a RRS loss of function (lof) animal, using the UAS-Gal4 system. The RRS-lof line was itself generated using CRISPR/Cas9 genome editing. Expression of both RRS WT and RRS SCR rescue the RRS-lof lethality. Adult animals expressing RRS WT and RRS SCR are compared and contrasted for their response to bacterial infection by gram positive M. luteus and gram negative Ecc15. We find that RRS SCR , when compared to RRS WT , shows modulation of the transcriptional response, as measured by quantitative 3' mRNA sequencing. Our study uncovers a possible non-canonical role for SUMOylation of RRS, a member of the MARS complex, in host-defense.
Collapse
Affiliation(s)
- Prajna Nayak
- Indian Institute of Science Education and Research (IISER), Pune, India
| | - Aarti Kejriwal
- Indian Institute of Science Education and Research (IISER), Pune, India
| | | |
Collapse
|
14
|
The role of SUMOylation during development. Biochem Soc Trans 2021; 48:463-478. [PMID: 32311032 PMCID: PMC7200636 DOI: 10.1042/bst20190390] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/17/2022]
Abstract
During the development of multicellular organisms, transcriptional regulation plays an important role in the control of cell growth, differentiation and morphogenesis. SUMOylation is a reversible post-translational process involved in transcriptional regulation through the modification of transcription factors and through chromatin remodelling (either modifying chromatin remodelers or acting as a ‘molecular glue’ by promoting recruitment of chromatin regulators). SUMO modification results in changes in the activity, stability, interactions or localization of its substrates, which affects cellular processes such as cell cycle progression, DNA maintenance and repair or nucleocytoplasmic transport. This review focuses on the role of SUMO machinery and the modification of target proteins during embryonic development and organogenesis of animals, from invertebrates to mammals.
Collapse
|
15
|
Luo Y, Fefelova E, Ninova M, Chen YCA, Aravin AA. Repression of interrupted and intact rDNA by the SUMO pathway in Drosophila melanogaster. eLife 2020; 9:e52416. [PMID: 33164748 PMCID: PMC7676866 DOI: 10.7554/elife.52416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/06/2020] [Indexed: 11/17/2022] Open
Abstract
Ribosomal RNAs (rRNAs) are essential components of the ribosome and are among the most abundant macromolecules in the cell. To ensure high rRNA level, eukaryotic genomes contain dozens to hundreds of rDNA genes, however, only a fraction of the rRNA genes seems to be active, while others are transcriptionally silent. We found that individual rDNA genes have high level of cell-to-cell heterogeneity in their expression in Drosophila melanogaster. Insertion of heterologous sequences into rDNA leads to repression associated with reduced expression in individual cells and decreased number of cells expressing rDNA with insertions. We found that SUMO (Small Ubiquitin-like Modifier) and SUMO ligase Ubc9 are required for efficient repression of interrupted rDNA units and variable expression of intact rDNA. Disruption of the SUMO pathway abolishes discrimination of interrupted and intact rDNAs and removes cell-to-cell heterogeneity leading to uniformly high expression of individual rDNA in single cells. Our results suggest that the SUMO pathway is responsible for both repression of interrupted units and control of intact rDNA expression.
Collapse
Affiliation(s)
- Yicheng Luo
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Elena Fefelova
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
- Institute of Molecular Genetics, Russian Academy of SciencesMoscowRussian Federation
| | - Maria Ninova
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Yung-Chia Ariel Chen
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Alexei A Aravin
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
16
|
Lin YL, Chung CL, Huang PJ, Chen CH, Fang SC. Revised annotation and extended characterizations of components of the Chlamydomonas reinhardtii SUMOylation system. PLANT DIRECT 2020; 4:e00266. [PMID: 33015534 PMCID: PMC7522501 DOI: 10.1002/pld3.266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 05/16/2023]
Abstract
Small ubiquitin-like modifier (SUMO) conjugation, or SUMOylation, is a reversible post-translational modification that is important for regulation of many cellular processes including cell division cycle in the eukaryotic kingdom. However, only a portion of the components of the Chlamydomonas SUMOylation system are known and their functions and regulation investigated. The present studies are aimed at extending discovery and characterization of new components and improving the annotation and nomenclature of all known proteins and genes involved in the system. Even though only one copy of the heterodimerized SUMO-activating enzyme, SAE1 and SAE2, was identified, the number of SUMO-conjugating enzymes (SCEs) and SUMO proteases/isopeptidase was expanded in Chlamydomonas. Using the reconstituted SUMOylation system, we showed that SCE1, SCE2, and SCE3 have SUMO-conjugating activity. In addition to SUMOylation, components required for other post-translational modifications such as NEDDylation, URMylation, and UFMylation, were confirmed to be present in Chlamydomonas. Our data also showed that besides isopeptidase activity, the SUMO protease domain of SUPPRESSOR OF MAT3 7/SENTRIN-SPECIFIC PROTEASE 1 (SMT7/SENP1) has endopeptidase activity that is capable of processing SUMO precursors. Moreover, the key cell cycle regulators of Chlamydomonas E2F1, DP1, CDKG1, CYCD2, and CYCD3 were SUMOylated in vitro, suggesting SUMOylation may be part of regulatory pathway modulating cell cycle regulators.
Collapse
Affiliation(s)
- Yen-Ling Lin
- Biotechnology Center in Southern Taiwan Academia Sinica Tainan Taiwan
- Agricultural Biotechnology Research Center Academia Sinica Taipei Taiwan
- Ph.D. Program in Microbial Genomics National Chung Hsing University and Academia Sinica Taichung Taiwan
| | - Chin-Lin Chung
- Biotechnology Center in Southern Taiwan Academia Sinica Tainan Taiwan
- Agricultural Biotechnology Research Center Academia Sinica Taipei Taiwan
| | - Pin-Jui Huang
- Biotechnology Center in Southern Taiwan Academia Sinica Tainan Taiwan
- Agricultural Biotechnology Research Center Academia Sinica Taipei Taiwan
| | - Chun-Han Chen
- Biotechnology Center in Southern Taiwan Academia Sinica Tainan Taiwan
- Agricultural Biotechnology Research Center Academia Sinica Taipei Taiwan
| | - Su-Chiung Fang
- Biotechnology Center in Southern Taiwan Academia Sinica Tainan Taiwan
- Agricultural Biotechnology Research Center Academia Sinica Taipei Taiwan
- Ph.D. Program in Microbial Genomics National Chung Hsing University and Academia Sinica Taichung Taiwan
- Institute of Tropical Plant Sciences and Microbiology National Cheng Kung University Tainan Taiwan
- National Cheng Kung University-Academia Sinica Graduate Program in Translational Agricultural Sciences Tainan Taiwan
| |
Collapse
|
17
|
Kaduskar B, Trivedi D, Ratnaparkhi GS. Caspar SUMOylation regulates Drosophila lifespan. MICROPUBLICATION BIOLOGY 2020; 2020. [PMID: 32760885 PMCID: PMC7396161 DOI: 10.17912/micropub.biology.000288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Deepti Trivedi
- Fly Facility, National Centre for Biological Sciences (NCBS), TIFR, Bangalore 560065 INDIA
| | | |
Collapse
|
18
|
Yau TY, Molina O, Courey AJ. SUMOylation in development and neurodegeneration. Development 2020; 147:147/6/dev175703. [PMID: 32188601 DOI: 10.1242/dev.175703] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In essentially all eukaryotes, proteins can be modified by the attachment of small ubiquitin-related modifier (SUMO) proteins to lysine side chains to produce branched proteins. This process of 'SUMOylation' plays essential roles in plant and animal development by altering protein function in spatially and temporally controlled ways. In this Primer, we explain the process of SUMOylation and summarize how SUMOylation regulates a number of signal transduction pathways. Next, we discuss multiple roles of SUMOylation in the epigenetic control of transcription. In addition, we evaluate the role of SUMOylation in the etiology of neurodegenerative disorders, focusing on Parkinson's disease and cerebral ischemia. Finally, we discuss the possibility that SUMOylation may stimulate survival and neurogenesis of neuronal stem cells.
Collapse
Affiliation(s)
- Tak-Yu Yau
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| | - Oscar Molina
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| | - Albert J Courey
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| |
Collapse
|
19
|
Hegde S, Soory A, Kaduskar B, Ratnaparkhi GS. SUMO conjugation regulates immune signalling. Fly (Austin) 2020; 14:62-79. [PMID: 32777975 PMCID: PMC7714519 DOI: 10.1080/19336934.2020.1808402] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
Post-translational modifications (PTMs) are critical drivers and attenuators for proteins that regulate immune signalling cascades in host defence. In this review, we explore functional roles for one such PTM, the small ubiquitin-like modifier (SUMO). Very few of the SUMO conjugation targets identified by proteomic studies have been validated in terms of their roles in host defence. Here, we compare and contrast potential SUMO substrate proteins in immune signalling for flies and mammals, with an emphasis on NFκB pathways. We discuss, using the few mechanistic studies that exist for validated targets, the effect of SUMO conjugation on signalling and also explore current molecular models that explain regulation by SUMO. We also discuss in detail roles of evolutionary conservation of mechanisms, SUMO interaction motifs, crosstalk of SUMO with other PTMs, emerging concepts such as group SUMOylation and finally, the potentially transforming roles for genome-editing technologies in studying the effect of PTMs.
Collapse
Affiliation(s)
- Sushmitha Hegde
- Biology, Indian Institute of Science Education & Research (IISER), Pune, India
| | - Amarendranath Soory
- Biology, Indian Institute of Science Education & Research (IISER), Pune, India
| | | | | |
Collapse
|
20
|
Ninova M, Chen YCA, Godneeva B, Rogers AK, Luo Y, Fejes Tóth K, Aravin AA. Su(var)2-10 and the SUMO Pathway Link piRNA-Guided Target Recognition to Chromatin Silencing. Mol Cell 2019; 77:556-570.e6. [PMID: 31901446 DOI: 10.1016/j.molcel.2019.11.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/11/2019] [Accepted: 11/08/2019] [Indexed: 01/14/2023]
Abstract
Regulation of transcription is the main mechanism responsible for precise control of gene expression. Whereas the majority of transcriptional regulation is mediated by DNA-binding transcription factors that bind to regulatory gene regions, an elegant alternative strategy employs small RNA guides, Piwi-interacting RNAs (piRNAs) to identify targets of transcriptional repression. Here, we show that in Drosophila the small ubiquitin-like protein SUMO and the SUMO E3 ligase Su(var)2-10 are required for piRNA-guided deposition of repressive chromatin marks and transcriptional silencing of piRNA targets. Su(var)2-10 links the piRNA-guided target recognition complex to the silencing effector by binding the piRNA/Piwi complex and inducing SUMO-dependent recruitment of the SetDB1/Wde histone methyltransferase effector. We propose that in Drosophila, the nuclear piRNA pathway has co-opted a conserved mechanism of SUMO-dependent recruitment of the SetDB1/Wde chromatin modifier to confer repression of genomic parasites.
Collapse
Affiliation(s)
- Maria Ninova
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA
| | - Yung-Chia Ariel Chen
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA
| | - Baira Godneeva
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA; Institute of Molecular Genetics, Russian Academy of Sciences, Moscow 123182, Russia
| | - Alicia K Rogers
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA
| | - Yicheng Luo
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA
| | - Katalin Fejes Tóth
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA.
| | - Alexei A Aravin
- California Institute of Technology, Division of Biology and Biological Engineering, 147-75, Pasadena, CA 91125, USA.
| |
Collapse
|
21
|
Rodriguez A, Briley SM, Patton BK, Tripurani SK, Rajapakshe K, Coarfa C, Rajkovic A, Andrieux A, Dejean A, Pangas SA. Loss of the E2 SUMO-conjugating enzyme Ube2i in oocytes during ovarian folliculogenesis causes infertility in mice. Development 2019; 146:dev.176701. [PMID: 31704792 PMCID: PMC6918767 DOI: 10.1242/dev.176701] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 10/29/2019] [Indexed: 01/25/2023]
Abstract
The number and quality of oocytes within the ovarian reserve largely determines fertility and reproductive lifespan in mammals. An oocyte-specific transcription factor cascade controls oocyte development, and some of these transcription factors, such as newborn ovary homeobox gene (NOBOX), are candidate genes for primary ovarian insufficiency in women. Transcription factors are frequently modified by the post-translational modification SUMOylation, but it is not known whether SUMOylation is required for function of the oocyte-specific transcription factors or if SUMOylation is required in oocytes during their development within the ovarian follicle. To test this, the sole E2 SUMO-conjugating enzyme, Ube2i, was ablated in mouse oocytes beginning in primordial follicles. Loss of oocyte Ube2i resulted in female infertility with major defects in stability of the primordial follicle pool, ovarian folliculogenesis, ovulation and meiosis. Transcriptomic profiling of ovaries suggests that loss of oocyte Ube2i caused defects in both oocyte- and granulosa cell-expressed genes, including NOBOX and some of its known target genes. Together, these studies show that SUMOylation is required in the mammalian oocyte during folliculogenesis for both oocyte development and communication with ovarian somatic cells.
Collapse
Affiliation(s)
- Amanda Rodriguez
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shawn M. Briley
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bethany K. Patton
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Swamy K. Tripurani
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kimal Rajapakshe
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aleksander Rajkovic
- Department of Pathology, University of California, San Francisco, CA 94134, USA
| | - Alexandra Andrieux
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Pasteur Institute, 75015 Paris, France
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Pasteur Institute, 75015 Paris, France
| | - Stephanie A. Pangas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA,Graduate Program in Biochemistry & Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA,Author for correspondence ()
| |
Collapse
|
22
|
Ubc9 deficiency selectively impairs the functionality of common lymphoid progenitors (CLPs) during bone marrow hematopoiesis. Mol Immunol 2019; 114:314-322. [PMID: 31442915 DOI: 10.1016/j.molimm.2019.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 10/26/2022]
Abstract
Hematopoietic development occurs in the bone marrow, and this process begins with hematopoietic stem cells (HSCs). Ubc9 is a unique E2-conjugating enzyme required for SUMOylation, an evolutionarily conserved post-translational modification system. We herein show that a conditional Ubc9 deletion in the hematopoietic system caused decreased thymus weight and reduced lymphocyte to myeloid cell ratio. Importantly, Ubc9 deletion in the hematopoietic system only selectively impaired the development of common lymphoid progenitors (CLPs) in the bone marrow and perturbed their potential to differentiate into lymphocytes, thereby decreasing the number of T/B cells in the periphery. Ubc9 was found to be required for CLP viability, and therefore, Ubc9 deficiency rendered CLPs to undergo apoptosis and attenuated their proliferation. Thus, Ubc9 plays a critical role in the regulation of CLP function during hematopoietic development in the bone marrow.
Collapse
|
23
|
Abrieu A, Liakopoulos D. How Does SUMO Participate in Spindle Organization? Cells 2019; 8:E801. [PMID: 31370271 PMCID: PMC6721559 DOI: 10.3390/cells8080801] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
The ubiquitin-like protein SUMO is a regulator involved in most cellular mechanisms. Recent studies have discovered new modes of function for this protein. Of particular interest is the ability of SUMO to organize proteins in larger assemblies, as well as the role of SUMO-dependent ubiquitylation in their disassembly. These mechanisms have been largely described in the context of DNA repair, transcriptional regulation, or signaling, while much less is known on how SUMO facilitates organization of microtubule-dependent processes during mitosis. Remarkably however, SUMO has been known for a long time to modify kinetochore proteins, while more recently, extensive proteomic screens have identified a large number of microtubule- and spindle-associated proteins that are SUMOylated. The aim of this review is to focus on the possible role of SUMOylation in organization of the spindle and kinetochore complexes. We summarize mitotic and microtubule/spindle-associated proteins that have been identified as SUMO conjugates and present examples regarding their regulation by SUMO. Moreover, we discuss the possible contribution of SUMOylation in organization of larger protein assemblies on the spindle, as well as the role of SUMO-targeted ubiquitylation in control of kinetochore assembly and function. Finally, we propose future directions regarding the study of SUMOylation in regulation of spindle organization and examine the potential of SUMO and SUMO-mediated degradation as target for antimitotic-based therapies.
Collapse
Affiliation(s)
- Ariane Abrieu
- CRBM, CNRS UMR5237, Université de Montpellier, 1919 route de Mende, 34090 Montpellier, France.
| | - Dimitris Liakopoulos
- CRBM, CNRS UMR5237, Université de Montpellier, 1919 route de Mende, 34090 Montpellier, France.
| |
Collapse
|
24
|
Bertke MM, Dubiak KM, Cronin L, Zeng E, Huber PW. A deficiency in SUMOylation activity disrupts multiple pathways leading to neural tube and heart defects in Xenopus embryos. BMC Genomics 2019; 20:386. [PMID: 31101013 PMCID: PMC6525467 DOI: 10.1186/s12864-019-5773-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/03/2019] [Indexed: 02/08/2023] Open
Abstract
Background Adenovirus protein, Gam1, triggers the proteolytic destruction of the E1 SUMO-activating enzyme. Microinjection of an empirically determined amount of Gam1 mRNA into one-cell Xenopus embryos can reduce SUMOylation activity to undetectable, but nonlethal, levels, enabling an examination of the role of this post-translational modification during early vertebrate development. Results We find that SUMOylation-deficient embryos consistently exhibit defects in neural tube and heart development. We have measured differences in gene expression between control and embryos injected with Gam1 mRNA at three developmental stages: early gastrula (immediately following the initiation of zygotic transcription), late gastrula (completion of the formation of the three primary germ layers), and early neurula (appearance of the neural plate). Although changes in gene expression are widespread and can be linked to many biological processes, three pathways, non-canonical Wnt/PCP, snail/twist, and Ets-1, are especially sensitive to the loss of SUMOylation activity and can largely account for the predominant phenotypes of Gam1 embryos. SUMOylation appears to generate different pools of a given transcription factor having different specificities with this post-translational modification involved in the regulation of more complex, as opposed to housekeeping, processes. Conclusions We have identified changes in gene expression that underlie the neural tube and heart phenotypes resulting from depressed SUMOylation activity. Notably, these developmental defects correspond to the two most frequently occurring congenital birth defects in humans, strongly suggesting that perturbation of SUMOylation, either globally or of a specific protein, may frequently be the origin of these pathologies. Electronic supplementary material The online version of this article (10.1186/s12864-019-5773-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michelle M Bertke
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA.,Present Address: College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Kyle M Dubiak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Laura Cronin
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Erliang Zeng
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana, 46556, USA.,Present Address: Division of Biostatistics and Computational Biology, Iowa Institute for Oral Health Research, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Preventive & Community Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Biostatistics, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA
| | - Paul W Huber
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA. .,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana, USA. .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, 46556, USA.
| |
Collapse
|
25
|
Michie KA, Bermeister A, Robertson NO, Goodchild SC, Curmi PMG. Two Sides of the Coin: Ezrin/Radixin/Moesin and Merlin Control Membrane Structure and Contact Inhibition. Int J Mol Sci 2019; 20:ijms20081996. [PMID: 31018575 PMCID: PMC6515277 DOI: 10.3390/ijms20081996] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 12/21/2022] Open
Abstract
The merlin-ERM (ezrin, radixin, moesin) family of proteins plays a central role in linking the cellular membranes to the cortical actin cytoskeleton. Merlin regulates contact inhibition and is an integral part of cell–cell junctions, while ERM proteins, ezrin, radixin and moesin, assist in the formation and maintenance of specialized plasma membrane structures and membrane vesicle structures. These two protein families share a common evolutionary history, having arisen and separated via gene duplication near the origin of metazoa. During approximately 0.5 billion years of evolution, the merlin and ERM family proteins have maintained both sequence and structural conservation to an extraordinary level. Comparing crystal structures of merlin-ERM proteins and their complexes, a picture emerges of the merlin-ERM proteins acting as switchable interaction hubs, assembling protein complexes on cellular membranes and linking them to the actin cytoskeleton. Given the high level of structural conservation between the merlin and ERM family proteins we speculate that they may function together.
Collapse
Affiliation(s)
- Katharine A Michie
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| | - Adam Bermeister
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| | - Neil O Robertson
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| | - Sophia C Goodchild
- Department of Molecular Sciences, Macquarie University, Sydney 2109, Australia.
| | - Paul M G Curmi
- School of Physics, University of New South Wales, Sydney 2052, Australia.
| |
Collapse
|
26
|
Morrell R, Sadanandom A. Dealing With Stress: A Review of Plant SUMO Proteases. FRONTIERS IN PLANT SCIENCE 2019; 10:1122. [PMID: 31620153 PMCID: PMC6759571 DOI: 10.3389/fpls.2019.01122] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/14/2019] [Indexed: 05/18/2023]
Abstract
The SUMO system is a rapid dynamic post-translational mechanism employed by eukaryotic cells to respond to stress. Plant cells experience hyperSUMOylation of substrates in response to stresses such as heat, ethanol, and drought. Many SUMOylated proteins are located in the nucleus, SUMOylation altering many nuclear processes. The SUMO proteases play two key functions in the SUMO cycle by generating free SUMO; they have an important role in regulating the SUMO cycle, and by cleaving SUMO off SUMOylated proteins, they provide specificity to which proteins become SUMOylated. This review summarizes the broad literature of plant SUMO proteases describing their catalytic activity, domains and structure, evolution, localization, and response to stress and highlighting potential new areas of research in the future.
Collapse
|
27
|
Umbaugh CS, Figueiredo ML. Lysines residing in putative Small Ubiquitin-like MOdifier (SUMO) motifs regulate fate and function of 37 KDa laminin receptor. Biochimie 2019; 156:92-99. [DOI: 10.1016/j.biochi.2018.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 10/06/2018] [Indexed: 01/17/2023]
|
28
|
Lim Y, Kim K, Lee Y. SUMOylation is required for fungal development and pathogenicity in the rice blast fungus Magnaporthe oryzae. MOLECULAR PLANT PATHOLOGY 2018; 19:2134-2148. [PMID: 29633464 PMCID: PMC6638150 DOI: 10.1111/mpp.12687] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 02/27/2018] [Accepted: 04/04/2018] [Indexed: 05/26/2023]
Abstract
Amongst the various post-translational modifications (PTMs), SUMOylation is a conserved process of attachment of a small ubiquitin-related modifier (SUMO) to a protein substrate in eukaryotes. This process regulates many important biological mechanisms, including transcriptional regulation, protein stabilization, cell cycle, DNA repair and pathogenesis. However, the functional role of SUMOylation is not well understood in plant-pathogenic fungi, including the model fungal pathogen Magnaporthe oryzae. In this study, we elucidated the roles of four SUMOylation-associated genes that encode one SUMO protein (MoSMT3), two E1 enzymes (MoAOS1 and MoUBA2) and one E2 enzyme (MoUBC9) in fungal development and pathogenicity. Western blot assays showed that SUMO modification was abolished in all deletion mutants. MoAOS1 and MoUBA2 were mainly localized in the nucleus, whereas MoSMT3 and MoUBC9 were localized in both the nucleus and cytoplasm. However, the four SUMOylation-associated proteins were predominantly localized in the nucleus under oxidative stress conditions. Deletion mutants for each of the four genes were viable, but showed significant defects in mycelial growth, conidiation, septum formation, conidial germination, appressorium formation and pathogenicity. Several proteins responsible for conidiation were predicted to be SUMOylated, suggesting that conidiation is controlled at the post-translational level by SUMOylation. In addition to infection-related development, SUMOylation also played important roles in resistance to nutrient starvation, DNA damage and oxidative stresses. Therefore, SUMOylation is required for infection-related fungal development, stress responses and pathogenicity in M. oryzae. This study provides new insights into the role of SUMOylation in the molecular mechanisms of pathogenesis of the rice blast fungus and other plant pathogens.
Collapse
Affiliation(s)
- You‐Jin Lim
- Department of Agricultural BiotechnologySeoul National UniversitySeoul 08826South Korea
| | - Ki‐Tae Kim
- Department of Agricultural BiotechnologySeoul National UniversitySeoul 08826South Korea
| | - Yong‐Hwan Lee
- Department of Agricultural BiotechnologySeoul National UniversitySeoul 08826South Korea
- Center for Fungal Genetic Resources, Plant Genomics and Breeding Institute, and Research Institute of Agriculture and Life Sciences, Seoul National UniversitySeoul 08826South Korea
| |
Collapse
|
29
|
Misra SK, Dighe K, Schwartz-Duval AS, Shang Z, Labriola LT, Pan D. In situ plasmonic generation in functional ionic-gold-nanogel scaffold for rapid quantitative bio-sensing. Biosens Bioelectron 2018; 120:77-84. [PMID: 30149216 DOI: 10.1016/j.bios.2018.08.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/21/2018] [Accepted: 08/10/2018] [Indexed: 11/17/2022]
Abstract
Conventional analytical techniques, which have been developed for high sensitivity and selectivity for the detection and quantification of relevant biomarkers, may not be as suitable for medical diagnosis in resource scarce environments as compared to point-of-care devices (POC). We have developed a new reactive sensing material which contains ionic gold entrapped within an agarose gel scaffold for POC quantification of ascorbic acid (AA) in tear fluid. Pathologically elevated concentration of AA in human tear fluid can serve as a biomarker for full-thickness injuries to the ocular surface, which are a medical emergency. This reactive sensing material will undergo colorimetric changes, quantitatively dependent on endogenous bio-reductants that are applied, as the entrapped ionic gold is reduced to form plasmonic nanoparticles. The capacity for this reactive material to function as a plasmonically driven biosensor, called 'OjoGel' (ojo-eye), was demonstrated with the endogenous reducing agent, AA. Through applications of AA of varied concentrations to the OjoGel, we demonstrated a quantitative colorimetric relationship between red (R) hexadecimal values and concentrations of AA in said treatments. This colorimetric relationship is directly resultant of plasmonic gold nanoparticle formation within the OjoGel scaffold. Using a commercially available mobile phone-based Pixel Picker® application, the OjoGel plasmonic sensing platform opens a new avenue for easy-to-use, rapid, and quantitative biosensing with low cost and accurate results.
Collapse
Affiliation(s)
- Santosh K Misra
- Department of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA; Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Ketan Dighe
- Department of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA; Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Aaron S Schwartz-Duval
- Department of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA; Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Zaixi Shang
- Department of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA; Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Leanne T Labriola
- Ophthalmology Department, Carle Foundation Hospital, Urbana, IL, USA; Surgery Department, University of Illinois College of Medicine, Urbana, IL, USA; Interdisciplinary Health Science Initiative, University of Illinois, Urbana, IL, USA
| | - Dipanjan Pan
- Department of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA; Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA; Carle Illinois College of Medicine, Urbana, IL, USA; Interdisciplinary Health Science Initiative, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
30
|
Wu J, Lei H, Zhang J, Chen X, Tang C, Wang W, Xu H, Xiao W, Gu W, Wu Y. Momordin Ic, a new natural SENP1 inhibitor, inhibits prostate cancer cell proliferation. Oncotarget 2018; 7:58995-59005. [PMID: 27449295 PMCID: PMC5312290 DOI: 10.18632/oncotarget.10636] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 07/08/2016] [Indexed: 12/13/2022] Open
Abstract
SUMO-specific protease 1 (SENP1), a member of the de-SUMOylation protease family, is elevated in prostate cancer (PCa) cells and is involved in PCa pathogenesis. Momordin Ιc (Mc), a natural pentacyclic triterpenoid, inhibited SENP1 in vitro, as reflected by reduced SENP1C-induced cleavage of SUMO2-ΔRanGAP1. Mc also altered the thermal stability of SENP1 in a newly developed cellular thermal shift assay, indicating that Mc directly interacts with SENP1 in PCa cells. Consistent with SENP1 inhibition, Mc increased SUMOylated protein levels, which was further confirmed by the accumulation of two known SUMOylated proteins, hypoxia inducible factor-1a and nucleus accumbens associated protein 1 in PC3 cells. Compared to LNCaP and normal prostate epithelial RWPE-1 cells, PC3 cells had higher levels of SENP1 mRNA and were more sensitive to Mc-induced growth inhibition. Mc also reduced SENP1 mRNA levels in PCa cells. Overexpression of SENP1 rescued PC3 cells from Mc-induced apoptosis. Finally, Mc suppressed cell proliferation and induced cell death in vivo in a xenograft PC3 tumor mouse model. These findings demonstrate that Mc is a novel SENP1 inhibitor with potential therapeutic value for PCa. Investigation of other pentacyclic triterpenoids may aid in the development of novel SENP1 inhibitor drugs.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of The Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinfu Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of The Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyun Chen
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caixia Tang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of The Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of The Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanzhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of The Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weilie Xiao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan, China
| | - Wenli Gu
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of The Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Comprehensive list of SUMO targets in Caenorhabditis elegans and its implication for evolutionary conservation of SUMO signaling. Sci Rep 2018; 8:1139. [PMID: 29348603 PMCID: PMC5773548 DOI: 10.1038/s41598-018-19424-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023] Open
Abstract
Post-translational modification by small ubiquitin-related modifier (SUMO) is a key regulator of cell physiology, modulating protein-protein and protein-DNA interactions. Recently, SUMO modifications were postulated to be involved in response to various stress stimuli. We aimed to identify the near complete set of proteins modified by SUMO and the dynamics of the modification in stress conditions in the higher eukaryote, Caenorhabditis elegans. We identified 874 proteins modified by SUMO in the worm. We have analyzed the SUMO modification in stress conditions including heat shock, DNA damage, arsenite induced cellular stress, ER and osmotic stress. In all these conditions the global levels of SUMOylation was significantly increased. These results show the evolutionary conservation of SUMO modifications in reaction to stress. Our analysis showed that SUMO targets are highly conserved throughout species. By comparing the SUMO targets among species, we approximated the total number of proteins modified in a given proteome to be at least 15–20%. We developed a web server designed for convenient prediction of potential SUMO modification based on experimental evidences in other species.
Collapse
|
32
|
The Biology of SUMO-Targeted Ubiquitin Ligases in Drosophila Development, Immunity, and Cancer. J Dev Biol 2018; 6:jdb6010002. [PMID: 29615551 PMCID: PMC5875560 DOI: 10.3390/jdb6010002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/27/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
The ubiquitin and SUMO (small ubiquitin-like modifier) pathways modify proteins that in turn regulate diverse cellular processes, embryonic development, and adult tissue physiology. These pathways were originally discovered biochemically in vitro, leading to a long-standing challenge of elucidating both the molecular cross-talk between these pathways and their biological importance. Recent discoveries in Drosophila established that ubiquitin and SUMO pathways are interconnected via evolutionally conserved SUMO-targeted ubiquitin ligase (STUbL) proteins. STUbL are RING ubiquitin ligases that recognize SUMOylated substrates and catalyze their ubiquitination, and include Degringolade (Dgrn) in Drosophila and RNF4 and RNF111 in humans. STUbL are essential for early development of both the fly and mouse embryos. In the fly embryo, Dgrn regulates early cell cycle progression, sex determination, zygotic gene transcription, segmentation, and neurogenesis, among other processes. In the fly adult, Dgrn is required for systemic immune response to pathogens and intestinal stem cell regeneration upon infection. These functions of Dgrn are highly conserved in humans, where RNF4-dependent ubiquitination potentiates key oncoproteins, thereby accelerating tumorigenesis. Here, we review the lessons learned to date in Drosophila and highlight their relevance to cancer biology.
Collapse
|
33
|
Choi BH, Chen C, Philips M, Dai W. RAS GTPases are modified by SUMOylation. Oncotarget 2017; 9:4440-4450. [PMID: 29435114 PMCID: PMC5796985 DOI: 10.18632/oncotarget.23269] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 11/19/2017] [Indexed: 01/31/2023] Open
Abstract
RAS proteins are GTPases that participate in multiple signal cascades, regulating crucial cellular processes including cell survival, proliferation, differentiation, and autophagy. Mutations or deregulated activities of RAS are frequently the driving force for oncogenic transformation and tumorigenesis. Given the important roles of the small ubiquitin-related modifier (SUMO) pathway in controlling the stability, activity, or subcellular localization of key cellular regulators, we investigated here whether RAS proteins are posttranslationally modified (i.e. SUMOylated) by the SUMO pathway. We observed that all three RAS protein isoforms (HRAS, KRAS, and NRAS) were modified by the SUMO3 protein. SUMOylation of KRAS protein, either endogenous or ectopically expressed, was observed in multiple cell lines. The SUMO3 modification of KRAS proteins could be removed by SUMO1/sentrin-specific peptidase 1 (SENP1) and SENP2, but not by SENP6, indicating that RAS SUMOylation is a reversible process. A conserved residue in RAS, Lys-42, was a site that mediates SUMOylation. Results from biochemical and molecular studies indicated that the SUMO-E3 ligase PIASγ specifically interacts with RAS and promotes its SUMOylation. Moreover, SUMOylation of RAS appeared to be associated with its activation. In summary, our study reveals a new posttranslational modification for RAS proteins. Since we found that HRAS, KRAS, and NRAS can all be SUMOylated, we propose that SUMOylation might represent a mechanism by which RAS activities are controlled.
Collapse
Affiliation(s)
- Byeong Hyeok Choi
- Department of Environmental Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Changyan Chen
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Mark Philips
- Department of Pathology, New York University Langone Medical Center, New York, NY, USA
| | - Wei Dai
- Department of Environmental Medicine, New York University Langone Medical Center, New York, NY, USA.,Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
34
|
Reversible regulation of ORC2 SUMOylation by PIAS4 and SENP2. Oncotarget 2017; 8:70142-70155. [PMID: 29050267 PMCID: PMC5642542 DOI: 10.18632/oncotarget.19594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/20/2017] [Indexed: 01/14/2023] Open
Abstract
The small ubiquitin-related modifier (SUMO) system is essential for smooth progression of cell cycle at the G2/M phase. Many centromeric proteins are reversibly SUMOylated to ensure proper chromosome segregation at the mitosis. SUMOylation of centromeric Origin Recognition Complex subunit 2 (ORC2) at the G2/M phase is essential in maintaining genome integrity. However, how ORC2 SUMOylation is regulated remains largely unclear. Here we show that ORC2 SUMOylation is reversibly controlled by SUMO E3 ligase PIAS4 and De-SUMOylase SENP2. Either depletion of PIAS4 or overexpression of SENP2 eliminated SUMOylation of ORC2 at the G/M phase and consequently resulted in abnormal centromeric histone H3 lysine 4 methylation. Cells stably expressing SENP2 protein or small interfering RNA for PIAS4 bypassed mitosis and endoreduplicated their genome to become polyploidy. Furthermore, percentage of polyploid cells is reduced after coexpression of ORC2-SUMO2 fusion protein. Thus, the proper regulation of ORC2 SUMOylation at the G2/M phase by PIAS4 and SENP2 is critical for smooth progression of the mitotic cycle of cells.
Collapse
|
35
|
Jox T, Buxa MK, Bohla D, Ullah I, Mačinković I, Brehm A, Bartkuhn M, Renkawitz R. Drosophila CP190- and dCTCF-mediated enhancer blocking is augmented by SUMOylation. Epigenetics Chromatin 2017; 10:32. [PMID: 28680483 PMCID: PMC5496309 DOI: 10.1186/s13072-017-0140-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/27/2017] [Indexed: 12/02/2022] Open
Abstract
Background Chromatin insulators shield promoters and chromatin domains from neighboring enhancers or chromatin regions with opposing activities. Insulator-binding proteins and their cofactors mediate the boundary function. In general, covalent modification of proteins by the small ubiquitin-like modifier (SUMO) is an important mechanism to control the interaction of proteins within complexes. Results Here we addressed the impact of dSUMO in respect of insulator function, chromatin binding of insulator factors and formation of insulator speckles in Drosophila. SUMOylation augments the enhancer blocking function of four different insulator sequences and increases the genome-wide binding of the insulator cofactor CP190. Conclusions These results indicate that enhanced chromatin binding of SUMOylated CP190 causes fusion of insulator speckles, which may allow for more efficient insulation. Electronic supplementary material The online version of this article (doi:10.1186/s13072-017-0140-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Theresa Jox
- Institute for Genetics, Justus-Liebig-University, 35392 Giessen, Germany.,Institute for Molecular Pathology, UKGM, 35392 Giessen, Germany
| | - Melanie K Buxa
- Institute for Genetics, Justus-Liebig-University, 35392 Giessen, Germany.,Flohr Consult, Adenauerallee 136, 53113 Bonn, Germany
| | - Dorte Bohla
- Institute for Genetics, Justus-Liebig-University, 35392 Giessen, Germany
| | - Ikram Ullah
- Institute of Molecular Biology and Tumour Research, Philipps University Marburg, 35037 Marburg, Germany
| | - Igor Mačinković
- Institute of Molecular Biology and Tumour Research, Philipps University Marburg, 35037 Marburg, Germany
| | - Alexander Brehm
- Institute of Molecular Biology and Tumour Research, Philipps University Marburg, 35037 Marburg, Germany
| | - Marek Bartkuhn
- Institute for Genetics, Justus-Liebig-University, 35392 Giessen, Germany
| | - Rainer Renkawitz
- Institute for Genetics, Justus-Liebig-University, 35392 Giessen, Germany
| |
Collapse
|
36
|
Nie M, Moser BA, Nakamura TM, Boddy MN. SUMO-targeted ubiquitin ligase activity can either suppress or promote genome instability, depending on the nature of the DNA lesion. PLoS Genet 2017; 13:e1006776. [PMID: 28475613 PMCID: PMC5438191 DOI: 10.1371/journal.pgen.1006776] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/19/2017] [Accepted: 04/24/2017] [Indexed: 11/19/2022] Open
Abstract
The posttranslational modifiers SUMO and ubiquitin critically regulate the DNA damage response (DDR). Important crosstalk between these modifiers at DNA lesions is mediated by the SUMO-targeted ubiquitin ligase (STUbL), which ubiquitinates SUMO chains to generate SUMO-ubiquitin hybrids. These SUMO-ubiquitin hybrids attract DDR proteins able to bind both modifiers, and/or are degraded at the proteasome. Despite these insights, specific roles for SUMO chains and STUbL in the DDR remain poorly defined. Notably, fission yeast defective in SUMO chain formation exhibit near wild-type resistance to genotoxins and moreover, have a greatly reduced dependency on STUbL activity for DNA repair. Based on these and other data, we propose that a critical role of STUbL is to antagonize DDR-inhibitory SUMO chain formation at DNA lesions. In this regard, we identify a SUMO-binding Swi2/Snf2 translocase called Rrp2 (ScUls1) as a mediator of the DDR defects in STUbL mutant cells. Therefore, in support of our proposal, SUMO chains attract activities that can antagonize STUbL and other DNA repair factors. Finally, we find that Taz1TRF1/TRF2-deficiency triggers extensive telomeric poly-SUMOylation. In this setting STUbL, together with its cofactor Cdc48p97, actually promotes genomic instability caused by the aberrant processing of taz1Δ telomeres by DNA repair factors. In summary, depending on the nature of the initiating DNA lesion, STUbL activity can either be beneficial or harmful. Since its discovery in 2007, SUMO-targeted ubiquitin ligase (STUbL) activity has been identified as a key regulator of diverse cellular processes such as DNA repair, mitosis and DNA replication. In each of these processes, STUbL has been shown to promote the chromatin extraction and/or degradation of SUMO chain modified proteins. However, it remains unclear whether STUbL acts as part of a "programmed" cascade to remove specific proteins, or antagonizes localized SUMO chain formation that otherwise impedes each process. Here we determine that SUMO chains, the major recruitment signal for STUbL, are largely dispensable for genotoxin resistance in fission yeast. Moreover, when SUMO chain formation is compromised, the need for STUbL activity in DNA repair is strongly reduced. These results indicate a primary role for STUbL in antagonizing localized SUMO chain formation. Interestingly, we also find that STUbL activity can be toxic at certain genomic lesions that induce extensive local SUMOylation. For example, STUbL promotes the chromosome instability and cell death caused by deprotected telomeres following Taz1TRF1/2 deletion. Together, our data suggest that STUbL limits DNA repair-inhibitory SUMO chain formation, and depending on the nature of the genomic lesion, can either suppress or cause genome instability.
Collapse
Affiliation(s)
- Minghua Nie
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States of America
| | - Bettina A. Moser
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Toru M. Nakamura
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Michael N. Boddy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States of America
- * E-mail:
| |
Collapse
|
37
|
Monribot-Villanueva J, Zurita M, Vázquez M. Developmental transcriptional regulation by SUMOylation, an evolving field. Genesis 2017; 55. [PMID: 27935206 DOI: 10.1002/dvg.23009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 02/05/2023]
Abstract
SUMOylation is a reversible post-translational protein modification that affects the intracellular localization, stability, activity, and interactions of its protein targets. The SUMOylation pathway influences several nuclear and cytoplasmic processes. The expression of many genes, in particular those involved in development is finely tuned in space and time by several groups of proteins. There is growing evidence that transcriptional regulation mechanisms involve direct SUMOylation of transcriptional-related proteins such as initiation and elongation factors, and subunits of chromatin modifier and remodeling complexes originally described as members of the trithorax and Polycomb groups in Drosophila. Therefore, it is being unveiled that SUMOylation has a role in both, gene silencing and gene activation mechanisms. The goal of this review is to discuss the information on how SUMO modification in components of these multi-subunit complexes may have an effect in genome architecture and function and, therefore, in the regulation of gene expression in time and space.
Collapse
Affiliation(s)
- Juan Monribot-Villanueva
- Departamento de Fisiología Molecular y Genética del Desarrollo, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Mario Zurita
- Departamento de Fisiología Molecular y Genética del Desarrollo, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Martha Vázquez
- Departamento de Fisiología Molecular y Genética del Desarrollo, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
38
|
Pirone L, Xolalpa W, Sigurðsson JO, Ramirez J, Pérez C, González M, de Sabando AR, Elortza F, Rodriguez MS, Mayor U, Olsen JV, Barrio R, Sutherland JD. A comprehensive platform for the analysis of ubiquitin-like protein modifications using in vivo biotinylation. Sci Rep 2017; 7:40756. [PMID: 28098257 PMCID: PMC5241687 DOI: 10.1038/srep40756] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/09/2016] [Indexed: 12/19/2022] Open
Abstract
Post-translational modification by ubiquitin and ubiquitin-like proteins (UbLs) is fundamental for maintaining protein homeostasis. Efficient isolation of UbL conjugates is hampered by multiple factors, including cost and specificity of reagents, removal of UbLs by proteases, distinguishing UbL conjugates from interactors, and low quantities of modified substrates. Here we describe bioUbLs, a comprehensive set of tools for studying modifications in Drosophila and mammals, based on multicistronic expression and in vivo biotinylation using the E. coli biotin protein ligase BirA. While the bioUbLs allow rapid validation of UbL conjugation for exogenous or endogenous proteins, the single vector approach can facilitate biotinylation of most proteins of interest. Purification under denaturing conditions inactivates deconjugating enzymes and stringent washes remove UbL interactors and non-specific background. We demonstrate the utility of the method in Drosophila cells and transgenic flies, identifying an extensive set of putative SUMOylated proteins in both cases. For mammalian cells, we show conjugation and localization for many different UbLs, with the identification of novel potential substrates for UFM1. Ease of use and the flexibility to modify existing vectors will make the bioUbL system a powerful complement to existing strategies for studying this important mode of protein regulation.
Collapse
Affiliation(s)
- Lucia Pirone
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Wendy Xolalpa
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Jón Otti Sigurðsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Juanma Ramirez
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Coralia Pérez
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Monika González
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | | | - Félix Elortza
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Manuel S Rodriguez
- ITAV, IPBS, Université de Toulouse, CNRS, UPS, 1 Place Pierre Potier Oncopole entrée B, BP 50624, 31106 Toulouse Cedex 1, France
| | - Ugo Mayor
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Alameda Urquijo, 36-5 Plaza Bizkaia, 48011 Bilbao, Spain
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Rosa Barrio
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - James D Sutherland
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| |
Collapse
|
39
|
Abstract
The ubiquitin -like protein SUMO is conjugated covalently to hundreds of target proteins in organisms throughout the eukaryotic domain. Genetic and biochemical studies using the model organism Drosophila melanogaster are beginning to reveal many essential functions for SUMO in cell biology and development. For example, SUMO regulates multiple signaling pathways such as the Ras/MAPK, Dpp, and JNK pathways. In addition, SUMO regulates transcription through conjugation to many transcriptional regulatory proteins, including Bicoid, Spalt , Scm, and Groucho. In some cases, conjugation of SUMO to a target protein inhibits its normal activity, while in other cases SUMO conjugation stimulates target protein activity. SUMO often modulates a biological process by altering the subcellular localization of a target protein. The ability of SUMO and other ubiquitin-like proteins to diversify protein function may be critical to the evolution of developmental complexity.
Collapse
|
40
|
Sumoylation in Development and Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:197-214. [DOI: 10.1007/978-3-319-50044-7_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Richard P, Vethantham V, Manley JL. Roles of Sumoylation in mRNA Processing and Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:15-33. [PMID: 28197904 DOI: 10.1007/978-3-319-50044-7_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SUMO has gained prominence as a regulator in a number of cellular processes. The roles of sumoylation in RNA metabolism, however, while considerable, remain less well understood. In this chapter we have assembled data from proteomic analyses, localization studies and key functional studies to extend SUMO's role to the area of mRNA processing and metabolism. Proteomic analyses have identified multiple putative sumoylation targets in complexes functioning in almost all aspects of mRNA metabolism, including capping, splicing and polyadenylation of mRNA precursors. Possible regulatory roles for SUMO have emerged in pre-mRNA 3' processing, where SUMO influences the functions of polyadenylation factors and activity of the entire complex. SUMO is also involved in regulating RNA editing and RNA binding by hnRNP proteins, and recent reports have suggested the involvement of the SUMO pathway in mRNA export. Together, these reports suggest that SUMO is involved in regulation of many aspects of mRNA metabolism and hold the promise for exciting future studies.
Collapse
Affiliation(s)
- Patricia Richard
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | | | - James L Manley
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
42
|
Di Genova BM, da Silva RC, da Cunha JPC, Gargantini PR, Mortara RA, Tonelli RR. Protein SUMOylation is Involved in Cell-cycle Progression and Cell Morphology in Giardia lamblia. J Eukaryot Microbiol 2016; 64:491-503. [PMID: 27864857 DOI: 10.1111/jeu.12386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/13/2016] [Accepted: 11/02/2016] [Indexed: 01/08/2023]
Abstract
The unicellular protozoa Giardia lamblia is a food- and waterborne parasite that causes giardiasis. This illness is manifested as acute and self-limited diarrhea and can evolve to long-term complications. Successful establishment of infection by Giardia trophozoites requires adhesion to host cells and colonization of the small intestine, where parasites multiply by mitotic division. The tight binding of trophozoites to host cells occurs by means of the ventral adhesive disc, a spiral array of microtubules and associated proteins such as giardins. In this work we show that knock down of the Small Ubiquitin-like MOdifier (SUMO) results in less adhesive trophzoites, decreased cell proliferation and deep morphological alterations, including at the ventral disc. Consistent with the reduced proliferation, SUMO knocked-down trophozoites were arrested in G1 and in S phases of the cell cycle. Mass spectrometry analysis of anti-SUMO immunoprecipitates was performed to identify SUMO substrates possibly involved in these events. Among the identified SUMOylation targets, α-tubulin was further validated by Western blot and confirmed to be a SUMO target in Giardia trophozoites.
Collapse
Affiliation(s)
- Bruno M Di Genova
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, RuaBotucatu 862, 04023-062, Vila Clementino, São Paulo, SP, Brazil
| | - Richard C da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, RuaBotucatu 862, 04023-062, Vila Clementino, São Paulo, SP, Brazil
| | - Júlia P C da Cunha
- Laboratório Especial de Ciclo Celular, Centro de Toxinas, Resposta Imune e Sinalização Celular - CeTICS, Instituto Butantan, Avenida Vital Brasil 1500, 05503-900, Butantã, São Paulo, SP, Brazil
| | - Pablo R Gargantini
- Laboratorio de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Católica de Córdoba (UCC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Armada Argentina 3555., X5016DHK, Cordoba, Argentina
| | - Renato A Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, RuaBotucatu 862, 04023-062, Vila Clementino, São Paulo, SP, Brazil
| | - Renata R Tonelli
- Departamento de Ciências Biológicas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Rua São Nicolau 210, 09913-030, Diadema, SP, Brazil
| |
Collapse
|
43
|
Xiao Y, Pollack D, Andrusier M, Levy A, Callaway M, Nieves E, Reddi P, Vigodner M. Identification of cell-specific targets of sumoylation during mouse spermatogenesis. Reproduction 2016; 151:149-66. [PMID: 26701181 DOI: 10.1530/rep-15-0239] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent findings suggest diverse and potentially multiple roles of small ubiquitin-like modifier (SUMO) in testicular function and spermatogenesis. However, SUMO targets remain uncharacterized in the testis due to the complex multicellular nature of testicular tissue, the inability to maintain and manipulate spermatogenesis in vitro, and the technical challenges involved in identifying low-abundance endogenous SUMO targets. In this study, we performed cell-specific identification of sumoylated proteins using concentrated cell lysates prepared with de-sumoylation inhibitors from freshly purified spermatocytes and spermatids. One-hundred and twenty proteins were uniquely identified in the spermatocyte and/or spermatid fractions. The identified proteins are involved in the regulation of transcription, stress response, microRNA biogenesis, regulation of major enzymatic pathways, nuclear-cytoplasmic transport, cell-cycle control, acrosome biogenesis, and other processes. Several proteins with important roles during spermatogenesis were chosen for further characterization by co-immunoprecipitation, co-localization, and in vitro sumoylation studies. GPS-SUMO Software was used to identify consensus and non-consensus sumoylation sites within the amino acid sequences of the proteins. The analyses confirmed the cell-specific sumoylation and/or SUMO interaction of several novel, previously uncharacterized SUMO targets such as CDK1, RNAP II, CDC5, MILI, DDX4, TDP-43, and STK31. Furthermore, several proteins that were previously identified as SUMO targets in somatic cells (KAP1 and MDC1) were identified as SUMO targets in germ cells. Many of these proteins have a unique role in spermatogenesis and during meiotic progression. This research opens a novel avenue for further studies of SUMO at the level of individual targets.
Collapse
Affiliation(s)
| | | | | | | | - Myrasol Callaway
- Department of BiologyStern College, Yeshiva University, New York, New York, USALaboratory for Macromolecular Analysis and ProteomicsAlbert Einstein College of Medicine, Bronx, New York, USADepartment of BiologyStern College, Yeshiva University, 245 Lexington Avenue, New York, New York 10016, USADepartment of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USADepartment of PathologyUniversity of Virginia, Charlottesville, Virginia, USA
| | - Edward Nieves
- Department of BiologyStern College, Yeshiva University, New York, New York, USALaboratory for Macromolecular Analysis and ProteomicsAlbert Einstein College of Medicine, Bronx, New York, USADepartment of BiologyStern College, Yeshiva University, 245 Lexington Avenue, New York, New York 10016, USADepartment of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USADepartment of PathologyUniversity of Virginia, Charlottesville, Virginia, USA
| | - Prabhakara Reddi
- Department of BiologyStern College, Yeshiva University, New York, New York, USALaboratory for Macromolecular Analysis and ProteomicsAlbert Einstein College of Medicine, Bronx, New York, USADepartment of BiologyStern College, Yeshiva University, 245 Lexington Avenue, New York, New York 10016, USADepartment of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USADepartment of PathologyUniversity of Virginia, Charlottesville, Virginia, USA
| | - Margarita Vigodner
- Department of BiologyStern College, Yeshiva University, New York, New York, USALaboratory for Macromolecular Analysis and ProteomicsAlbert Einstein College of Medicine, Bronx, New York, USADepartment of BiologyStern College, Yeshiva University, 245 Lexington Avenue, New York, New York 10016, USADepartment of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USADepartment of PathologyUniversity of Virginia, Charlottesville, Virginia, USA Department of BiologyStern College, Yeshiva University, New York, New York, USALaboratory for Macromolecular Analysis and ProteomicsAlbert Einstein College of Medicine, Bronx, New York, USADepartment of BiologyStern College, Yeshiva University, 245 Lexington Avenue, New York, New York 10016, USADepartment of Developmental and Molecular BiologyAlbert Einstein College of Medicine, Bronx, New York, USADepartment of PathologyUniversity of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
44
|
Xiao Y, Lucas B, Molcho E, Schiff T, Vigodner M. Inhibition of CDK1 activity by sumoylation. Biochem Biophys Res Commun 2016; 478:919-23. [PMID: 27520372 DOI: 10.1016/j.bbrc.2016.08.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Abstract
Sumoylation (a covalent modification by Small Ubiquitin-like Modifiers or SUMO proteins) has been implicated in the regulation of various cellular events including cell cycle progression. We have recently identified CDK1, a master regulator of mitosis and meiosis, as a SUMO target both in vivo and in vitro, supporting growing evidence concerning a close cross talk between sumoylation and phosphorylation during cell cycle progression. However, any data regarding the effect of sumoylation upon CDK1 activity have been missing. In this study, we performed a series of in vitro experiments to inhibit sumoylation by three different means (ginkgolic acid, physiological levels of oxidative stress, and using an siRNA approach) and assessed the changes in CDK1 activity using specific antibodies and a kinase assay. We have also tested for an interaction between SUMO and active and/or inactive CDK1 isoforms in addition to having assessed the status of CDK1-interacting sumoylated proteins upon inhibition of sumoylation. Our data suggest that inhibition of sumoylation increases the activity of CDK1 probably through changes in sumoylated status and/or the ability of specific proteins to bind CDK1 and inhibit its activity.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Biology, Stern College, Yeshiva University, New York, NY, USA
| | - Benjamin Lucas
- Department of Biology, Stern College, Yeshiva University, New York, NY, USA
| | - Elana Molcho
- Department of Biology, Stern College, Yeshiva University, New York, NY, USA
| | - Tania Schiff
- Department of Biology, Stern College, Yeshiva University, New York, NY, USA
| | - Margarita Vigodner
- Department of Biology, Stern College, Yeshiva University, New York, NY, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
45
|
Kariithi HM, Boeren S, Murungi EK, Vlak JM, Abd-Alla AMM. A proteomics approach reveals molecular manipulators of distinct cellular processes in the salivary glands of Glossina m. morsitans in response to Trypanosoma b. brucei infections. Parasit Vectors 2016; 9:424. [PMID: 27485005 PMCID: PMC4969678 DOI: 10.1186/s13071-016-1714-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/20/2016] [Indexed: 12/28/2022] Open
Abstract
Background Glossina m. morsitans is the primary vector of the Trypanosoma brucei group, one of the causative agents of African trypanosomoses. The parasites undergo metacyclogenesis, i.e. transformation into the mammalian-infective metacyclic trypomastigote (MT) parasites, in the salivary glands (SGs) of the tsetse vector. Since the MT-parasites are largely uncultivable in vitro, information on the molecular processes that facilitate metacyclogenesis is scanty. Methods To bridge this knowledge gap, we employed tandem mass spectrometry to investigate protein expression modulations in parasitized (T. b. brucei-infected) and unparasitized SGs of G. m. morsitans. We annotated the identified proteins into gene ontologies and mapped the up- and downregulated proteins within protein-protein interaction (PPI) networks. Results We identified 361 host proteins, of which 76.6 % (n = 276) and 22.3 % (n = 81) were up- and downregulated, respectively, in parasitized SGs compared to unparasitized SGs. Whilst 32 proteins were significantly upregulated (> 10-fold), only salivary secreted adenosine was significantly downregulated. Amongst the significantly upregulated proteins, there were proteins associated with blood feeding, immunity, cellular proliferation, homeostasis, cytoskeletal traffic and regulation of protein turnover. The significantly upregulated proteins formed major hubs in the PPI network including key regulators of the Ras/MAPK and Ca2+/cAMP signaling pathways, ubiquitin-proteasome system and mitochondrial respiratory chain. Moreover, we identified 158 trypanosome-specific proteins, notable of which were proteins in the families of the GPI-anchored surface glycoproteins, kinetoplastid calpains, peroxiredoxins, retrotransposon host spot multigene and molecular chaperones. Whilst immune-related trypanosome proteins were over-represented, membrane transporters and proteins involved in translation repression (e.g. ribosomal proteins) were under-represented, potentially reminiscent of the growth-arrested MT-parasites. Conclusions Our data implicate the significantly upregulated proteins as manipulators of diverse cellular processes in response to T. b. brucei infection, potentially to prepare the MT-parasites for invasion and evasion of the mammalian host immune defences. We discuss potential strategies to exploit our findings in enhancement of trypanosome refractoriness or reduce the vector competence of the tsetse vector. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1714-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Henry M Kariithi
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O Box 57811, 00200, Kaptagat Rd, Loresho, Nairobi, Kenya. .,Insect Pest Control Laboratories, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Wagrammer Straße 5, Vienna, Austria.
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University, Dreijenlaan 3, 6703, HA, Wageningen, The Netherlands
| | - Edwin K Murungi
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, 20115, Njoro, Kenya
| | - Just M Vlak
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708, PB, Wageningen, The Netherlands
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratories, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Wagrammer Straße 5, Vienna, Austria.
| |
Collapse
|
46
|
Wang X, Yu Q, Huang L, Yu P. Lentivirus-mediated inhibition of USP39 suppresses the growth of gastric cancer cells via PARP activation. Mol Med Rep 2016; 14:301-306. [PMID: 27175747 DOI: 10.3892/mmr.2016.5252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 03/10/2016] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer (GC) is the second most common cause of cancer-associated mortality worldwide. Ubiquitin-specific peptidase 39 (USP39) has important roles in mRNA processing and has been reported to be involved in the growth of breast cancer cells. However, the roles of USP39 in GC have remained to be investigated, which was the aim of the present study. A lentivirus expressing short hairpin RNA targeting USP39 was constructed and transfected into MGC80‑3 cells. Suppression of USP39 expression significantly decreased the proliferation and colony forming ability of MGC80‑3 cells as indicated by an MTT and a clonogenic assay, respectively. In addition, flow cytometric cell cycle analysis revealed that depression of USP39 induced G2/M‑phase arrest, while an intracellular signaling array showed that the cleavage of PARP at Asp214 was increased following USP39 knockdown. These results suggested that USP39 is involved in the proliferation of GCs and may be utilized as a molecular target for GC therapy.
Collapse
Affiliation(s)
- Xinbao Wang
- Department of Abdominal Tumor Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Qiming Yu
- Department of Abdominal Tumor Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Ling Huang
- Department of Abdominal Tumor Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Pengfei Yu
- Department of Abdominal Tumor Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
47
|
Meng F, Qian J, Yue H, Li X, Xue K. SUMOylation of Rb enhances its binding with CDK2 and phosphorylation at early G1 phase. Cell Cycle 2016; 15:1724-32. [PMID: 27163259 PMCID: PMC4957593 DOI: 10.1080/15384101.2016.1182267] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Retinoblastoma protein (Rb) is a prototypical tumor suppressor that is vital to the negative regulation of the cell cycle and tumor progression. Hypo-phosphorylated Rb is associated with G0/G1 arrest by suppressing E2F transcription factor activity, whereas Rb hyper-phosphorylation allows E2F release and cell cycle progression from G0/G1 to S phase. However, the factors that regulate cyclin-dependent protein kinase (CDK)-dependent hyper-phosphorylation of Rb during the cell cycle remain obscure. In this study, we show that throughout the cell cycle, Rb is specifically small ubiquitin-like modifier (SUMO)ylated at early G1 phase. SUMOylation of Rb stimulates its phosphorylation level by recruiting a SUMO-interaction motif (SIM)-containing kinase CDK2, leading to Rb hyper-phosphorylation and E2F-1 release. In contrast, a SUMO-deficient Rb mutant results in reduced SUMOylation and phosphorylation, weakened CDK2 binding, and attenuated E2F-1 sequestration. Furthermore, we reveal that Rb SUMOylation is required for cell proliferation. Therefore, our study describes a novel mechanism that regulates Rb phosphorylation during cell cycle progression.
Collapse
Affiliation(s)
- Fengxi Meng
- a Department of Ophthalmology , Eye and ENT Hospital of Fudan University , Shanghai , China.,b Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University , Shanghai , China
| | - Jiang Qian
- a Department of Ophthalmology , Eye and ENT Hospital of Fudan University , Shanghai , China.,b Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University , Shanghai , China
| | - Han Yue
- a Department of Ophthalmology , Eye and ENT Hospital of Fudan University , Shanghai , China.,b Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University , Shanghai , China
| | - Xiaofeng Li
- a Department of Ophthalmology , Eye and ENT Hospital of Fudan University , Shanghai , China.,b Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University , Shanghai , China
| | - Kang Xue
- a Department of Ophthalmology , Eye and ENT Hospital of Fudan University , Shanghai , China.,b Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University , Shanghai , China
| |
Collapse
|
48
|
Abstract
This review discusses our current understanding of the small ubiquitin-like modifier (SUMO) pathway and how it functionally intersects with Ras signaling in cancer. The Ras family of small GTPases are frequently mutated in cancer. The role of the SUMO pathway in cancer and in Ras signaling is currently not well understood. Recent studies have shown that the SUMO pathway can both regulate Ras/MAPK pathway activity directly and support Ras-driven oncogenesis through the regulation of proteins that are not direct Ras effectors. We recently discovered that in Ras mutant cancer cells, the SUMOylation status of a subset of proteins is altered and one such protein, KAP1, is required for Ras-driven transformation. A better understanding of the functional interaction between the SUMO and Ras pathways could lead to new insights into the mechanism of Ras-driven oncogenesis.
Collapse
Affiliation(s)
- Haibo Zhang
- a Laboratory of Canter Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH , Bethesda , MD , USA
| | - Ji Luo
- a Laboratory of Canter Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH , Bethesda , MD , USA
| |
Collapse
|
49
|
Lv X, Pan C, Zhang Z, Xia Y, Chen H, Zhang S, Guo T, Han H, Song H, Zhang L, Zhao Y. SUMO regulates somatic cyst stem cells maintenance and directly targets hedgehog pathway in adult Drosophila testis. Development 2016; 143:1655-62. [DOI: 10.1242/dev.130773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/16/2016] [Indexed: 01/12/2023]
Abstract
SUMO (Small ubiquitin-related modifier) modification (SUMOylation) is a highly dynamic post-translational modification (PTM) playing important roles in tissue development and disease progression. However, its function in adult stem cell maintenance is largely unknown. Here we report the function of SUMOylation in somatic cyst stem cells (CySCs) self-renewal in adult Drosophila testis. The SUMO pathway cell-autonomously regulates CySCs maintenance. Reduction of SUMOylation promotes premature differentiation of CySCs and impedes the proliferation of CySCs, which finally reduce the number of CySCs. Consistently, CySC clones carrying mutation of the SUMO conjugating enzyme are rapidly lost. Furthermore, inhibition of SUMO pathway phenocopies the disruption of Hedgehog (Hh) pathway, and can block the promoted proliferation of CySCs by Hh activation. Importantly, SUMO pathway directly regulates the SUMOylation of Hh pathway transcriptional factor, Cubitus interruptus (Ci), which is required for promoting CySCs proliferation. Thus, we conclude that SUMO directly targets Hh pathway and regulates CySCs maintenance in adult Drosophila testis.
Collapse
Affiliation(s)
- Xiangdong Lv
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Chenyu Pan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Zhao Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Yuanxin Xia
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Hao Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Shuo Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Tong Guo
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Hui Han
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Haiyun Song
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of sciences, Shanghai 200031, P.R. China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, P.R. China
| | - Yun Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, P.R. China
| |
Collapse
|
50
|
Pirone L, Xolalpa W, Mayor U, Barrio R, Sutherland JD. Analysis of SUMOylated Proteins in Cells and In Vivo Using the bioSUMO Strategy. Methods Mol Biol 2016; 1475:161-9. [PMID: 27631805 DOI: 10.1007/978-1-4939-6358-4_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Posttranslational regulation of proteins by conjugation of ubiquitin- and ubiquitin-like molecules is a common theme in almost every known biological pathway. SUMO (small ubiquitin-related modifier) is dynamically added and deleted from many cellular substrates to control activity, localization, and recruitment of other SUMO-recognizing protein complexes. The dynamic nature of this modification and its low abundance in resting cells make it challenging to study, with susceptibility to deSUMOylases further complicating its analysis. Here we describe bioSUMO, a general method to isolate and analyze SUMOylated proteins from cultured cells, using Drosophila as a highlighted example. The method also has been validated in transgenic flies, as well as human cells. SUMOylated substrates are labeled by in vivo biotinylation, which facilitates their subsequent purification using streptavidin-based affinity chromatography under stringent conditions and with very low background. The bioSUMO approach can be used to validate whether a specific protein is modified, or used to analyze an entire SUMO subproteome. If coupled to quantitative proteomics methods, it may reveal how the SUMO landscape changes with different stimuli, or in diverse cell or tissue types. This technique offers a complementary approach to study SUMO biology and we expect that the strategy can be extended to other ubiquitin-like proteins.
Collapse
Affiliation(s)
- Lucia Pirone
- CIC bioGUNE, Bizkaia Technology Park, Derio, Bizkaia, 48160, Spain
| | - Wendy Xolalpa
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, 62100, Mexico
| | - Ugo Mayor
- Biokimika eta Biologia Molekularra Saila, Zientzia eta Teknologia Fakultatea, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, 48940, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, 48013, Spain
| | - Rosa Barrio
- CIC bioGUNE, Bizkaia Technology Park, Derio, Bizkaia, 48160, Spain.
| | | |
Collapse
|