201
|
Abstract
Derivation of induced Pluripotent Stem Cells (iPSCs) by reprogramming somatic cells to a pluripotent state has revolutionized stem cell research. Ensuing this, various groups have used genetic and non-genetic approaches to generate iPSCs from numerous cell types. However, achieving a pluripotent state in most of the reprogramming studies is marred by serious limitations such as low reprogramming efficiency and slow kinetics. These limitations are mainly due to the presence of potent barriers that exist during reprogramming when a mature cell is coaxed to achieve a pluripotent state. Several studies have revealed that intrinsic factors such as non-optimal stoichiometry of reprogramming factors, specific signaling pathways, cellular senescence, pluripotency-inhibiting transcription factors and microRNAs act as a roadblock. In addition, the epigenetic state of somatic cells and specific epigenetic modifications that occur during reprogramming also remarkably impede the generation of iPSCs. In this review, we present a comprehensive overview of the barriers that inhibit reprogramming and the understanding of which will pave the way to develop safe strategies for efficient reprogramming.
Collapse
|
202
|
SUMOylation Targets Adeno-associated Virus Capsids but Mainly Restricts Transduction by Cellular Mechanisms. J Virol 2020; 94:JVI.00871-20. [PMID: 32669341 DOI: 10.1128/jvi.00871-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/08/2020] [Indexed: 11/20/2022] Open
Abstract
Adeno-associated virus (AAV) has proven to be a promising candidate for gene therapy due to its nonpathogenic nature, ease of production, and broad tissue tropism. However, its transduction capabilities are not optimal due to the interaction with various host factors within the cell. In a previous study, we identified members of the small ubiquitin-like modifier (SUMO) pathway as significant restriction factors in AAV gene transduction. In the present study, we explored the scope of this restriction by focusing on the AAV capsid and host cell proteins as targets. We show that during vector production, the capsid protein VP2 becomes SUMOylated, as indicated by deletion and point mutations of VP2 or the obstruction of its N terminus via the addition of a tag. We observed that SUMOylated AAV capsids display higher stability than non-SUMOylated capsids. Prevention of capsid SUMOylation by VP2 mutations did not abolish transduction restriction by SUMOylation; however, it reduced activation of gene transduction by shutdown of the cellular SUMOylation pathway. This indicates a link between capsid SUMOylation and SUMOylation of cellular proteins in restricting gene transduction. Infection with AAV triggers general SUMOylation of cellular proteins. In particular, the DAXX protein, a putative host cell restriction factor that can become SUMOylated, is able to restrict AAV gene transduction by reducing the intracellular accumulation of AAV vectors. We also observe that the coexpression of a SUMOylation inhibitor with an AAV2 reporter gene vector increased gene transduction significantly.IMPORTANCE Host factors within the cell are the major mode of restriction of adeno-associated virus (AAV) and keep it from fulfilling its maximum potential as a gene therapy vector. A better understanding of the intricacies of restriction would enable the engineering of better vectors. Via a genome-wide short interfering RNA screen, we identified that proteins of the small ubiquitin-like modifier (SUMO) pathway play an important role in AAV restriction. In this study, we investigate whether this restriction is targeted to the AAV directly or indirectly through host cell factors. The results indicate that both targets act in concert to restrict AAV.
Collapse
|
203
|
Xiao Q, Chen XH, Jiang RC, Chen SY, Chen KF, Zhu X, Zhang XL, Huang JJ, Qin Y, Zhang GP, Yi Q, Luo JD. Ubc9 Attenuates Myocardial Ischemic Injury Through Accelerating Autophagic Flux. Front Pharmacol 2020; 11:561306. [PMID: 33041800 PMCID: PMC7522513 DOI: 10.3389/fphar.2020.561306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/14/2020] [Indexed: 01/09/2023] Open
Abstract
Aims SUMOylation is a post-translational modification that plays a crucial role in the cellular stress response. We aimed to demonstrate whether and how the SUMO E2 conjugation enzyme Ubc9 affects acute myocardial ischemic (MI) injury. Methods and Results Adenovirus expressing Ubc9 was administrated by multipoint injection in the border zone of heart immediately after MI in C57BL/6 mice. Neonatal rat cardiomyocytes (NRCMs) were also infected, followed by oxygen and glucose deprivation (OGD). In vivo, Ubc9 adenovirus-injected mice showed decreased cardiomyocyte apoptosis, reduced myocardial fibrosis, and improved cardiac function post-MI. In vitro, overexpression of Ubc9 decreased cardiomyocyte apoptosis, whereas silence of Ubc9 showed the opposite results during OGD. We next found that Ubc9 significantly decreased the accumulation of autophagy marker p62/SQSTM, while the LC3 II level hardly changed. When in the presence of bafilomycin A1 (BAF), the Ubc9 adenovirus plus OGD group presented a higher level of LC3 II and GFP-LC3 puncta than the OGD group. Moreover, the Ubc9 adenovirus group displayed increased numbers of yellow plus red puncta and a rising ratio of red to yellow puncta on the mRFP-GFP-LC3 fluorescence assay, indicating that Ubc9 induces an acceleration of autophagic flux from activation to degradation. Mechanistically, Ubc9 upregulated SUMOylation of the core proteins Vps34 and Beclin1 in the class III phosphatidylinositol 3-kinase (PI3K-III) complexes and boosted the protein assembly of PI3K-III complex I and II under OGD. Moreover, the colocalization of Vps34 with autophagosome marker LC3 or lysosome marker Lamp1 was augmented after Ubc9 overexpression, indicating a positive effect of Ubc9-boosted protein assembly of the PI3K-III complexes on autophagic flux enhancement. Conclusions We uncovered a novel role of Ubc9 in protecting cardiomyocytes from ischemic stress via Ubc9-induced SUMOylation, leading to increased PI3K-III complex assembly and autophagy-positioning. These findings may indicate a potential therapeutic target, Ubc9, for treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Qing Xiao
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacology, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiu-Hui Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Ru-Chao Jiang
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Sheng-Ying Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Kai-Feng Chen
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Xiang Zhu
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Ling Zhang
- Department of Neonatology, Maternal and Children Hospital of Guangdong Province, Guangzhou, China
| | - Jun-Jun Huang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan Qin
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Gui-Ping Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Quan Yi
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacology, Guangzhou Medical University, Guangzhou, China
| | - Jian-Dong Luo
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacology, Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
204
|
Wang K, Jiang Z, Lu X, Zhang Y, Yuan X, Luo D, Lin Z, Zuo Y, Luo Q. Cardiomyocyte-specific deletion of Senp2 contributes to CVB3 viral replication and inflammation. Int Immunopharmacol 2020; 88:106941. [PMID: 33182061 DOI: 10.1016/j.intimp.2020.106941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 10/23/2022]
Abstract
Viral myocarditis (VMC) is characterized by cardiac inflammation and excessive inflammatory responses after viral infection. SENP2, a deSUMO-specific protease, has been reported to regulate antiviral innate immunity. This study aimed to investigate whether SENP2 affects CVB3-induced VMC. We generated a CVB3-induced VMC mouse model in 6-week-old cardiomyocyte-specific Senp2 knockout mice. The mice were sacrificed at days 0, 2, 4 and 6 after CVB3 infection. The survival rate, body weight, myocardial histopathological changes, viral load, cytokine levels and antiviral gene expression in cardiac tissues of both groups were investigated. Our study indicated that the expression of Senp2 in primary cardiomyocytes was upregulated by CVB3 infection. Moreover, deletion of Senp2 in the heart exacerbated CVB3 infection-induced myocarditis, facilitated CVB3 viral replication and downregulated the expression of antiviral proteins. In conclusion, our findings suggest a protective role for SENP2 in CVB3-induced VMC.
Collapse
Affiliation(s)
- Kangwei Wang
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Zaixue Jiang
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Xiaomei Lu
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Yaozhong Zhang
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Xu Yuan
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Dong Luo
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Zitian Lin
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China
| | - Yong Zuo
- The Department of Biochemistry and Molecular & Cell Biology, The Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medical, Shanghai 200025, China.
| | - Qingming Luo
- Department of Children's Genetics and Infectious Diseases Laboratory, Dongguan Institute of Pediatrics, Dongguan, Guangdong 510000, China; Department of Respiratory Medicine, Dongguan Children's Hospital, Dongguan, Guangdong 510000, China.
| |
Collapse
|
205
|
Liu YC, Hsu WL, Ma YL, Lee EHY. Melatonin Induction of APP Intracellular Domain 50 SUMOylation Alleviates AD through Enhanced Transcriptional Activation and Aβ Degradation. Mol Ther 2020; 29:376-395. [PMID: 32950104 PMCID: PMC7791018 DOI: 10.1016/j.ymthe.2020.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/06/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
The amyloid precursor protein (APP) intracellular domain (AICD) is implicated in the pathogenesis of Alzheimer’s disease (AD), but post-translational modification of AICD has rarely been studied and its role in AD is unknown. In this study, we examined the role and molecular mechanism of AICD SUMOylation in the pathogenesis of AD. We found that AICD is SUMO-modified by the SUMO E3 ligase protein inhibitor of activated STAT1 (PIAS1) in the hippocampus at Lys-43 predominantly, and that knockdown of PIAS1 decreases endogenous AICD SUMOylation. AICD SUMOylation increases AICD association with its binding protein Fe65 and increases AICD nuclear translocation. Furthermore, AICD SUMOylation increases AICD association with cyclic AMP-responsive element binding protein (CREB) and p65 and their DNA binding for transcriptional activation of neprilysin (NEP) and transthyretin (TTR), two major Aβ-degrading enzymes, respectively. Consequently, AICD SUMOylation decreases the Aβ level, Aβ oligomerization, and amyloid plaque deposits. It also rescues spatial memory deficits in APP/PS1 mice. Conversely, blockade of AICD SUMOylation at Lys-43 produces the opposite effects. Melatonin is identified as an endogenous stimulus that induces AICD SUMOylation. It also decreases the Aβ level and rescues reduction of PIAS1, NEP, and TTR expression in APP/PS1 mice. In this study, we demonstrate that AICD SUMOylation functions as a novel endogenous defense mechanism to combat AD.
Collapse
Affiliation(s)
- Yen-Chen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Wei-Lun Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yun-Li Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Eminy H Y Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.
| |
Collapse
|
206
|
Li S, Wang J, Hu G, Aman S, Li B, Li Y, Xia K, Yang Y, Ahmad B, Wang M, Wu H. SUMOylation of MCL1 protein enhances its stability by regulating the ubiquitin-proteasome pathway. Cell Signal 2020; 73:109686. [DOI: 10.1016/j.cellsig.2020.109686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
|
207
|
Li C, Sharma-Bhandari A, Seo JH, Kim Y. Lysyl oxidase-variant 2 (LOX-v2) colocalizes with promyelocytic leukemia-nuclear bodies in the nucleus. IUBMB Life 2020; 72:2400-2408. [PMID: 32852148 DOI: 10.1002/iub.2363] [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: 05/27/2020] [Revised: 07/01/2020] [Accepted: 07/18/2020] [Indexed: 01/30/2023]
Abstract
Lysyl oxidase-variant 2 (LOX-v2) is a novel variant of LOX that functions as an amine oxidase for the formation of collagen and elastin fibrils in the extracellular matrix (ECM). LOX-v2 lacks the N-terminal prepropeptide region of LOX but contains the C-terminal domains required for amine oxidase activity. To study the cellular localization of LOX-v2, we generated a recombinant construct of LOX-v2 with an epitope tag at the C-terminus and then transfected the recombinant construct into HEK293 cells. Upon ectopic expression, LOX-v2 showed much higher expression in the nucleus than in the cytoplasm. In coimmunofluorescence staining with subnuclear structures, LOX-v2 colocalized with the promyelocytic leukemia-nuclear bodies (PML-NBs). Further, the ectopic expression of LOX-v2 increased global SUMOylation in the nucleus. PML-NBs have been implicated in various cellular activities, including transcriptional regulation, DNA repair, cell cycle control, anti-viral response, and apoptosis. Our findings strongly indicate that LOX-v2 may be subject to different cellular processing from what LOX undergoes, playing a distinct functional role in the PML-NBs, beyond the cross-linking of the structural proteins.
Collapse
Affiliation(s)
- Chunying Li
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Anjali Sharma-Bhandari
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Jae Ho Seo
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Youngho Kim
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Republic of Korea
| |
Collapse
|
208
|
Kitagawa T, Takiya S. Regulation of genes for ubiquitination and SUMO‐specific protease involved in larval development of the silkworm,
Bombyx mori. Dev Growth Differ 2020; 62:438-449. [DOI: 10.1111/dgd.12687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/26/2020] [Accepted: 05/30/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Tsuyuki Kitagawa
- Graduate School of Life Science Hokkaido University Sapporo Japan
| | - Shigeharu Takiya
- Graduate School of Life Science Hokkaido University Sapporo Japan
- Division of Biological Sciences and Center for Genome Dynamics Faculty of Science Hokkaido University Sapporo Japan
| |
Collapse
|
209
|
Genomic Analysis of the SUMO-Conjugating Enzyme and Genes under Abiotic Stress in Potato ( Solanum tuberosum L.). Int J Genomics 2020; 2020:9703638. [PMID: 32802829 PMCID: PMC7335410 DOI: 10.1155/2020/9703638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/24/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
SUMO-conjugating enzymes (SCE) and SUMO (Small Ubiquitin-Like Modifiers) genes are important components of SUMOylation. SCE has a crucial role during the SUMOylation process which acts as a catalyst to transfer SUMO to the target protein. Comprehensive studies on SCE and SUMO have been performed in some plants, but studies on these genes remain limited in potato. This study is aimed at exploring the role of StSCE and StSUMO genes in abiotic stress conditions. Nine and seven putative StSCEs and StSUMO genes, respectively, were identified using different methods and databases available for potato. Chromosomal localization showed that SCE and StSUMO genes are unevenly distributed on 7 different chromosomes. Potato genome database was accessed for the expression profile of StSCE and StSUMO genes, and these genes were differentially expressed in different tissues and organs during different phases of plant growth. The expression patterns on different treatments were further evaluated using qRT-PCR for all the StSCE and StSUMO genes. The expression was upregulated in StSCE1/5/6 and 7 under salt and PEG treatment. StSUMO 1/2 and 4 were upregulated under salt stress whereas StSCE9 and StSUMO2 and 4 were observed downregulated under PEG treatment. The results of this study could be useful to explore the role of StSCE genes in potato improvement.
Collapse
|
210
|
Du L, Fakih MG, Rosen ST, Chen Y. SUMOylation of E2F1 Regulates Expression of EZH2. Cancer Res 2020; 80:4212-4223. [PMID: 32816857 DOI: 10.1158/0008-5472.can-20-1259] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/19/2020] [Accepted: 07/28/2020] [Indexed: 11/16/2022]
Abstract
Elevated expression of EZH2, the enzymatic subunit of polycomb repressive complex 2 (PRC2), often occurs in cancer. EZH2 expression results in the silencing of genes that suppress tumor formation and metastasis through trimethylation of histone H3 at lysine 27 (H3K27me3) at their promoters. However, inhibitors of EZH2 enzymatic activity have not shown the expected efficacy against cancer in clinical trials, suggesting a need for other strategies to address EZH2 overexpression. Here, we show that SUMOylation, a posttranslational modification characterized by covalent attachment of small ubiquitin-like modifier (SUMO) proteins to a lysine (Lys) residue on target proteins, enhances EZH2 transcription. Either knockdown of the SUMO-activating enzyme SAE2 or pharmacologic inhibition of SUMOylation resulted in decreased levels of EZH2 mRNA and protein as well as reduced H3K27me3 levels. SUMOylation regulated EZH2 expression by enhancing binding of the E2F1 transcriptional activator to the EZH2 promoter. Inhibition of SUMOylation not only resulted in reduced EZH2 mRNA and protein levels but also increased expression of genes silenced by EZH2, such as E-cadherin, which suppresses epithelial-mesenchymal transition and metastasis. In more than 6,500 patient tumor samples across different cancer types, expression of UBA2 and EZH2 was positively correlated. Taken together, our findings suggest that inhibition of SUMOylation may serve as a potential strategy to address EZH2 overexpression and improve current cancer therapeutic approaches. SIGNIFICANCE: These findings provide important biological insights into the mechanism of EZH2 overexpression in cancers and suggest that inhibiting SUMOylation may improve current cancer therapeutic approaches.
Collapse
Affiliation(s)
- Li Du
- Department of Molecular Medicine, City of Hope, Duarte, California.,Toni Stephenson Lymphoma Center, Beckman Research Institute of City of Hope, Duarte, California
| | - Marwan G Fakih
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, California
| | - Steven T Rosen
- Beckman Research Institute and Comprehensive Cancer Center, City of Hope, Duarte, California.
| | - Yuan Chen
- Department of Surgery and Moores Cancer Center, UC San Diego Health, La Jolla, California.
| |
Collapse
|
211
|
Yu F, Zhang W, Yan C, Yan D, Zhou M, Chen J, Zhao X, Zhu A, Zhou J, Liu H, Sun H, Fu Y. PAX6, modified by SUMOylation, plays a protective role in corneal endothelial injury. Cell Death Dis 2020; 11:683. [PMID: 32826860 PMCID: PMC7442823 DOI: 10.1038/s41419-020-02848-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 11/21/2022]
Abstract
Treating corneal endothelial diseases tends to be challenging as human corneal endothelial cells (CECs) do not proliferate in vivo. The pathogenesis or mechanisms underlying injured CECs need further studies. The abnormal expression of PAX6, which is an essential transcription factor for corneal homeostasis, exhibits corneal endothelial defects. However, the effects of PAX6 protein involved in corneal endothelial wound process are still unknown. Here, we found the upregulated protein levels of PAX6 in human corneal endothelial monolayer after injury; the expression of PAX6 also increased in murine and rat corneal endothelium injury models. Enforced PAX6 expression could alleviate the damages to CECs via regulating permeability by prompting cellular tight junction. In addition, SUMOylation mainly happened on both K53 and K89 residues of 48-kD PAX6 (the longest and main isoform expressed in cornea), and de-SUMOylation promoted the stability of PAX6 protein in vitro. In CECs of SENP1+/− mice, increased SUMOylation levels leading to instability and low expression of PAX6, delayed the repair of CECs after injury. Furthermore, overexpression of PAX6 accelerated the rate of corneal endothelial repair of SENP1+/− mice. Our findings indicate that SENP1-mediated de-SUMOylation improving the stability of PAX6, amplifies the protective effects of PAX6 on corneal endothelial injuries, highlighting potentials of PAX6 and/or SUMOylation to be used as a treatment target for corneal endothelial disorders.
Collapse
Affiliation(s)
- Fei Yu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Weijie Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Chenxi Yan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Dan Yan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Meng Zhou
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Junzhao Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Xiangteng Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Aoxue Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huiqing Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Department of Pediatric Neurosurgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hao Sun
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China.
| | - Yao Fu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China.
| |
Collapse
|
212
|
He J, Cheng J, Wang T. SUMOylation-Mediated Response to Mitochondrial Stress. Int J Mol Sci 2020; 21:ijms21165657. [PMID: 32781782 PMCID: PMC7460625 DOI: 10.3390/ijms21165657] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial stress is considered as a factor that reprograms the mitochondrial biogenesis and metabolism. As known, SUMOylation occurs through a series of stress-induced biochemical reactions. During the process of SUMOylation, the small ubiquitin-like modifier (SUMO) and its specific proteases (SENPs) are key signal molecules. Furthermore, they are considered as novel mitochondrial stress sensors that respond to the signals produced by various stresses. The responses are critical for mitochondrial homeostasis. The scope of this review is to provide an overview of the function of SUMOylation in the mitochondrial stress response, to delineate a SUMOylation-involved signal network diagram, and to highlight a number of key questions that remain answered.
Collapse
Affiliation(s)
- Jianli He
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (J.C.); (T.W.); Tel.: +86-(21)-6384-6590-776327 (J.C.); +86-(21)-6384-6590-778026 (T.W.)
| | - Tianshi Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (J.C.); (T.W.); Tel.: +86-(21)-6384-6590-776327 (J.C.); +86-(21)-6384-6590-778026 (T.W.)
| |
Collapse
|
213
|
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
|
214
|
Chen C, Sun X, Xie W, Chen S, Hu Y, Xing D, Xu J, Chen X, Zhao Z, Han Z, Xue X, Shen X, Lin K. Opposing biological functions of the cytoplasm and nucleus DAXX modified by SUMO-2/3 in gastric cancer. Cell Death Dis 2020; 11:514. [PMID: 32641734 PMCID: PMC7343808 DOI: 10.1038/s41419-020-2718-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
Death domain-associated protein (DAXX) is a complex biological multifunctional protein and is involved in the tumorigenesis and progression of multiple cancers. The accumulation of DAXX in the nucleus is a common phenomenon in tumor cells. However, altering the subcellular localizations of DAXX results in different biological functions, and we also found that its nuclear/cytoplasmic ratio (NCR) was associated with poor prognosis in gastric cancer (GC). In this study, we investigated the effect of cytoplasmic and nuclear DAXX (cDAXX and nDAXX) in GC and the underlying mechanisms. Immunohistochemical detection performed in 323 GC tissues reveled that cDAXX was associated with a better survival, while high nDAXX expression suggested a poorer prognosis outcome. Upregulation of DAXX in the cytoplasm inhibited cell proliferation and promoted apoptosis, whereas downregulation of DAXX in the nucleus displayed opposite effects. Moreover, Transwell assays revealed that DAXX enhanced GC cell migration and invasion. Analysis from the Gene Expression Profile Interactive Analysis (GEPIA) database showed that the expression of DAXX was significantly associated with SUMO-2/3 in GC tissues. Co-immunoprecipitation combined with immunofluorescence analysis indicated that DAXX interacted directly with SUMO-2/3. Subsequently, down-regulating the expression of SUMO-2/3 resulted in altered subcellular localization of DAXX. Bioinformatics analysis showed that RanBP2 may act as SUMO E3 ligase to promote nuclear-plasma transport via combining with RanGAP1. Taken together, our results indicated that DAXX plays opposing roles in GC and suggest a new model whereby cDAXX, nDAXX, and SUMO-2/3 form a molecular network that regulates the subcellular localization of DAXX and thereby modulates its opposing biological effects. Thus, our findings provide a foundation for future studies of DAXX as a novel therapeutic target for patients with GC.
Collapse
Affiliation(s)
- Chenbin Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Xiangwei Sun
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Wangkai Xie
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Sian Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Yuanbo Hu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Dong Xing
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Jianfeng Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Xiaodong Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Zhiguang Zhao
- Department of Pathology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Zheng Han
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China.
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China.
| | - Kezhi Lin
- Morphology Laboratory, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, P.R. China.
| |
Collapse
|
215
|
Zeidler M. Fine-Tuning by the Touch of Big Dudes: SUMOs Role in Light Signaling. MOLECULAR PLANT 2020; 13:943-945. [PMID: 32522624 DOI: 10.1016/j.molp.2020.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 06/11/2023]
Affiliation(s)
- Mathias Zeidler
- Institute of Plant Physiology, Justus-Liebig-University Giessen, Senckenbergstrasse 3, 35390 Giessen, Germany.
| |
Collapse
|
216
|
SUMOylation stabilizes hSSB1 and enhances the recruitment of NBS1 to DNA damage sites. Signal Transduct Target Ther 2020; 5:80. [PMID: 32576812 PMCID: PMC7311467 DOI: 10.1038/s41392-020-0172-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
Human single-stranded DNA-binding protein 1 (hSSB1) is required for the efficient recruitment of the MRN complex to DNA double-strand breaks and is essential for the maintenance of genome integrity. However, the mechanism by which hSSB1 recruits NBS1 remains elusive. Here, we determined that hSSB1 undergoes SUMOylation at both K79 and K94 under normal conditions and that this modification is dramatically enhanced in response to DNA damage. SUMOylation of hSSB1, which is specifically fine-tuned by PIAS2α, and SENP2, not only stabilizes the protein but also enhances the recruitment of NBS1 to DNA damage sites. Cells with defective hSSB1 SUMOylation are sensitive to ionizing radiation, and global inhibition of SUMOylation by either knocking out UBC9 or adding SUMOylation inhibitors significantly enhances the sensitivity of cancer cells to etoposide. Our findings reveal that SUMOylation, as a novel posttranslational modification of hSSB1, is critical for the functions of this protein, indicating that the use of SUMOylation inhibitors (e.g., 2-D08 and ML-792) may be a new strategy that would benefit cancer patients being treated with chemo- or radiotherapy.
Collapse
|
217
|
Rijkschroeff P, Schoenmaker T, Caspers M, Verschuren L, Keijser BJF, Nicu EA, Loos BG. Dentistry and OMICS: Transcriptome Dynamics of an Oral Ecosystem as Measured by Changes in Oral Polymorphonuclear Neutrophils in Experimental Gingivitis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:531-540. [PMID: 32559408 DOI: 10.1089/omi.2020.0034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Oral health and dentistry are essential components of systems medicine, which has received lesser attention in comparison to other medical fields, such as cancer biology. In this context, oral polymorphonuclear neutrophils (oPMNs) play an important role in the maintenance of oral health. To the best of our knowledge, this is the first study to report original observations on the transcriptional responses of oPMNs during experimentally induced gingivitis, by temporarily refraining from regular oral care. Oral rinses were prospectively collected at four different time points for oPMNs isolation from healthy volunteers: day 1 (start of the experimental gingivitis challenge), day 9 (during challenge), day 14 (end of the challenge), and day 21 (postchallenge). Transcriptome of oPMNs was determined by RNA sequencing. Differentially expressed genes (DEGs) were selected at p < 0.01 level, and evaluated for pathway regulation using Ingenuity Pathway Analysis suite. We found four major clusters of DEGs, consisting of 256 initial response DEGs (day 9 only), 221 late response DEGs (day 14 only), 53 persistent responsive DEGs (consistent at day 9 and 14), and 524 DEGs showing responses only in the postchallenge phase (day 21 only). Pathway analysis of the initial and late response DEGs showed involvement in many immune regulatory pathways and PMN function, whereas DEGs at day 21 were associated with epithelial adherence signaling and other miscellaneous related signaling pathways. The results from this pilot study showed that oPMNs mediate oral inflammatory processes, suggesting their immunomodulatory role in oral equilibrium.
Collapse
Affiliation(s)
- Patrick Rijkschroeff
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UVA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UVA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands
| | - Martien Caspers
- Microbiology and System Biology, TNO Earth, Environmental and Life Sciences, Zeist, The Netherlands
| | - Lars Verschuren
- Microbiology and System Biology, TNO Earth, Environmental and Life Sciences, Zeist, The Netherlands
| | - Bart J F Keijser
- Microbiology and System Biology, TNO Earth, Environmental and Life Sciences, Zeist, The Netherlands.,Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Elena A Nicu
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UVA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands.,Opris Dent SRL, Sibiu, Romania
| | - Bruno G Loos
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam (UVA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands
| |
Collapse
|
218
|
Nakka VP, Mohammed AQ. A Critical Role for ISGylation, Ubiquitination and, SUMOylation in Brain Damage: Implications for Neuroprotection. Neurochem Res 2020; 45:1975-1985. [DOI: 10.1007/s11064-020-03066-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/12/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
|
219
|
Sun M, Chen X, Yin YX, Gao Y, Zhang L, Chen B, Ji Y, Fukunaga K, Han F, Lu YM. Role of pericyte-derived SENP1 in neuronal injury after brain ischemia. CNS Neurosci Ther 2020; 26:815-828. [PMID: 32495523 PMCID: PMC7366739 DOI: 10.1111/cns.13398] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/13/2020] [Accepted: 04/26/2020] [Indexed: 12/21/2022] Open
Abstract
Aims SUMOylation is a posttranslational modification related to multiple human diseases. SUMOylation can be reversed by classes of proteases known as the sentrin/SUMO‐specific proteases (SENPs). In the present study, we investigate the potential role of SENP1 in pericytes in the brain ischemia. Methods Pericyte‐specific deletion of senp1 mice (Cspg4‐Cre; senp1f/f) were used for brain function and neuronal damage evaluation following brain ischemia. The cerebral blood vessels of diameter, velocity, and flux were performed in living mice by two‐photon laser scanning microscopy (TPLSM). Biochemical analysis and immunohistochemistry methods were used to address the role and mechanism of pericyte‐specific SENP1 in the pathological process of brain ischemia. A coculture model of HBVPs and HBMECs mimicked the BBB in vitro and was used to evaluate BBB integrity after glucose deprivation. Results Our results showed that senp1‐specific deletion in pericytes did not affect the motor function and cognitive function of mice. However, the pericyte‐specific deletion of senp1 aggravated the infarct size and motor deficit following focal brain ischemia. Consistently, the TPLSM data demonstrated that SENP1 deletion in pericytes accelerated thrombosis formation in brain microvessels. We also found that pericyte‐specific deletion of senp1 exaggerated the neuronal damage significantly following brain ischemia in mice. Moreover, SENP1 knockdown in pericytes could activate the apoptosis signaling and disrupt the barrier integrity in vitro coculture model. Conclusions Our findings revealed that targeting SENP1 in pericytes may represent a novel therapeutic strategy for neurovascular protection in stroke.
Collapse
Affiliation(s)
- Meiling Sun
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiang Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yi-Xuan Yin
- Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yinping Gao
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Li Zhang
- Department of Geriatrics, Nanjing Brain Hospital affiliated to Nanjing Medical University, Nanjing, China
| | - Boqian Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yin Ji
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Simcere Pharmaceutical Group, Nanjing, China
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Ying-Mei Lu
- Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
220
|
Mechanisms and roles of mitochondrial localisation and dynamics in neuronal function. Neuronal Signal 2020; 4:NS20200008. [PMID: 32714603 PMCID: PMC7373250 DOI: 10.1042/ns20200008] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 01/23/2023] Open
Abstract
Neurons are highly polarised, complex and incredibly energy intensive cells, and their demand for ATP during neuronal transmission is primarily met by oxidative phosphorylation by mitochondria. Thus, maintaining the health and efficient function of mitochondria is vital for neuronal integrity, viability and synaptic activity. Mitochondria do not exist in isolation, but constantly undergo cycles of fusion and fission, and are actively transported around the neuron to sites of high energy demand. Intriguingly, axonal and dendritic mitochondria exhibit different morphologies. In axons mitochondria are small and sparse whereas in dendrites they are larger and more densely packed. The transport mechanisms and mitochondrial dynamics that underlie these differences, and their functional implications, have been the focus of concerted investigation. Moreover, it is now clear that deficiencies in mitochondrial dynamics can be a primary factor in many neurodegenerative diseases. Here, we review the role that mitochondrial dynamics play in neuronal function, how these processes support synaptic transmission and how mitochondrial dysfunction is implicated in neurodegenerative disease.
Collapse
|
221
|
Olig2 SUMOylation protects against genotoxic damage response by antagonizing p53 gene targeting. Cell Death Differ 2020; 27:3146-3161. [PMID: 32483381 PMCID: PMC7560653 DOI: 10.1038/s41418-020-0569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 01/05/2023] Open
Abstract
Posttranslational modifications of nuclear proteins, including transcription factors, nuclear receptors, and their coregulators, have attracted much attention in cancer research. Although phosphorylation of oligodendrocyte transcription factor 2 (Olig2) may contribute to the notorious resistance of gliomas to radiation and genotoxic drugs, the precise mechanisms remain elusive. We show here that in addition to phosphorylation, Olig2 is also conjugated by small ubiquitin-like modifier-1 (SUMO1) at three lysine residues K27, K76, and K112. SUMOylation is required for Olig2 to suppress p53-mediated cell cycle arrest and apoptosis induced by genotoxic damage, and to enhance resistance to temozolomide (TMZ) in glioma. Both SUMOylation and triple serine motif (TSM) phosphorylation of Olig2 are required for the antiapoptotic function. Olig2 SUMOylation enhances its genetic targeting ability, which in turn occludes p53 recruitment to Cdkn1a promoter for DNA-damage responses. Our work uncovers a SUMOylation-dependent regulatory mechanism of Olig2 in regulating cancer survival.
Collapse
|
222
|
Lin X, Wang Y, Jiang Y, Xu M, Pang Q, Sun J, Yu Y, Shen Z, Lei R, Xu J. Sumoylation enhances the activity of the TGF-β/SMAD and HIF-1 signaling pathways in keloids. Life Sci 2020; 255:117859. [PMID: 32474020 DOI: 10.1016/j.lfs.2020.117859] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 12/29/2022]
Abstract
Excessive fibrosis and extracellular matrix deposition resulting from upregulation of target genes expression mediated by transforming growth factor-beta (TGF-β)/SMAD and hypoxia inducible factor-1 (HIF-1) signaling pathways are the main mechanisms that drive keloid formation. Sumoylation is a protein posttranslational modification that regulates the function of proteins in many biological processes. In the present study, we aimed to investigate the mechanism underlying the effects of sumoylation on the TGF-β/SMAD and HIF-1 signaling pathways in keloids. We used 2-D08 to block sumoylation and silenced the expression of sentrin sumo-specific protease 1 (SENP1) to enhance sumoylation in human foreskin fibroblasts (HFFs) and human keloid fibroblasts (HKFs). We also reduced and increased intracellular SUMO1 levels by silencing SUMO1 and transfecting cells with a SUMO1 overexpression lentivirus, respectively. Sumoylation has the ability to amplify TGF-β/SMAD and HIF-1 signals in keloids, while SUMO1, especially the SUMO1-RanGAP1 complex, is the key molecule affecting the TGF-β/SMAD and HIF-1 signaling pathways. In addition, we also found that hypoxia promotes sumoylation in keloids and that HIF-1α is covalently modified by SUMO1 at Lys 391 and Lys 477 in HKFs. In summary, we elucidated the role and molecular mechanism of sumoylation in the formation of keloids, providing a new perspective for a potential therapeutic target of keloids.
Collapse
Affiliation(s)
- Xiaohu Lin
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yuming Wang
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yan Jiang
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Mingyuan Xu
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qianqian Pang
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jiaqi Sun
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yijia Yu
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Zeren Shen
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Rui Lei
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jinghong Xu
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
223
|
Paul S, Ainavarapu SRK, Venkatramani R. Variance of Atomic Coordinates as a Dynamical Metric to Distinguish Proteins and Protein-Protein Interactions in Molecular Dynamics Simulations. J Phys Chem B 2020; 124:4247-4262. [PMID: 32281802 DOI: 10.1021/acs.jpcb.0c01191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Protein dynamics is a manifestation of the complex trajectories of these biomolecules on a multidimensional rugged potential energy surface (PES) driven by thermal energy. At present, computational methods such as atomistic molecular dynamics (MD) simulations can describe thermal protein conformational changes in fully solvated environments over millisecond timescales. Despite these advances, a quantitative assessment of protein dynamics remains a complicated topic, intricately linked to issues such as sampling convergence and the identification of appropriate reaction coordinates/structural features to describe protein conformational states and motions. Here, we present the cumulative variance of atomic coordinate fluctuations (CVCF) along trajectories as an intuitive PES sensitive metric to assess both the extent of sampling and protein dynamics captured in MD simulations. We first examine the sampling problem in model one- (1D) and two-dimensional (2D) PES to demonstrate that the CVCF when traced as a function of the sampling variable (time in MD simulations) can identify local and global equilibria. Further, even far from global equilibrium, a situation representative of standard MD trajectories of proteins, the CVCF can distinguish different PES and therefore resolve the resultant protein dynamics. We demonstrate the utility of our CVCF analysis by applying it to distinguish the dynamics of structurally homologous proteins from the ubiquitin family (ubiquitin, SUMO1, SUMO2) and ubiquitin protein-protein interactions. Our CVCF analysis reveals that differential side-chain dynamics from the structured part of the protein (the conserved β-grasp fold) present distinct protein PES to distinguish ubiquitin from SUMO isoforms. Upon binding to two functionally distinct protein partners (UBCH5A and UEV), intrinsic ubiquitin dynamics changes to reflect the binding context even though the two proteins have similar binding modes, which lead to negligible (sub-angstrom scale) structural changes.
Collapse
Affiliation(s)
- Sanjoy Paul
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Dr. Homi Bhabha Road, Colaba, Mumbai 400005, Maharashtra, India
| | - Sri Rama Koti Ainavarapu
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Dr. Homi Bhabha Road, Colaba, Mumbai 400005, Maharashtra, India
| | - Ravindra Venkatramani
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Dr. Homi Bhabha Road, Colaba, Mumbai 400005, Maharashtra, India
| |
Collapse
|
224
|
Ge Z, Li A, McNamara J, Dos Remedios C, Lal S. Pathogenesis and pathophysiology of heart failure with reduced ejection fraction: translation to human studies. Heart Fail Rev 2020; 24:743-758. [PMID: 31209771 DOI: 10.1007/s10741-019-09806-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Heart failure represents the end result of different pathophysiologic processes, which culminate in functional impairment. Regardless of its aetiology, the presentation of heart failure usually involves symptoms of pump failure and congestion, which forms the basis for clinical diagnosis. Pathophysiologic descriptions of heart failure with reduced ejection fraction (HFrEF) are being established. Most commonly, HFrEF is centred on a reactive model where a significant initial insult leads to reduced cardiac output, further triggering a cascade of maladaptive processes. Predisposing factors include myocardial injury of any cause, chronically abnormal loading due to hypertension, valvular disease, or tachyarrhythmias. The pathophysiologic processes behind remodelling in heart failure are complex and reflect systemic neurohormonal activation, peripheral vascular effects and localised changes affecting the cardiac substrate. These abnormalities have been the subject of intense research. Much of the translational successes in HFrEF have come from targeting neurohormonal responses to reduced cardiac output, with blockade of the renin-angiotensin-aldosterone system (RAAS) and beta-adrenergic blockade being particularly fruitful. However, mortality and morbidity associated with heart failure remains high. Although systemic neurohormonal blockade slows disease progression, localised ventricular remodelling still adversely affects contractile function. Novel therapy targeted at improving cardiac contractile mechanics in HFrEF hold the promise of alleviating heart failure at its source, yet so far none has found success. Nevertheless, there are increasing calls for a proximal, 'cardiocentric' approach to therapy. In this review, we examine HFrEF therapy aimed at improving cardiac function with a focus on recent trials and emerging targets.
Collapse
Affiliation(s)
- Zijun Ge
- Sydney Medical School, University of Sydney, Camperdown, Australia
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Amy Li
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
- Department of Pharmacy and Biomedical Science, La Trobe University, Melbourne, Australia
| | - James McNamara
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Cris Dos Remedios
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Sean Lal
- Sydney Medical School, University of Sydney, Camperdown, Australia.
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
- Cardiac Research Laboratory, Discipline of Anatomy and Histology, University of Sydney, Anderson Stuart Building (F13), Camperdown, NSW, 2006, Australia.
| |
Collapse
|
225
|
Zhang F, Zhang W. Encrypting Chemical Reactivity in Protein Sequences toward
Information‐Coded
Reactions
†. CHINESE J CHEM 2020. [DOI: 10.1002/cjoc.202000083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Fan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Wen‐Bin Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| |
Collapse
|
226
|
Carnino JM, Ni K, Jin Y. Post-translational Modification Regulates Formation and Cargo-Loading of Extracellular Vesicles. Front Immunol 2020; 11:948. [PMID: 32528471 PMCID: PMC7257894 DOI: 10.3389/fimmu.2020.00948] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022] Open
Abstract
Accumulating evidence suggests that post-translational modifications (PTMs) regulate the selective encapsulation of non-coding RNA molecules into extracellular vesicles (EVs) and contribute to the downstream functions of EVs or EV-cargo non-coding RNAs. EVs are a newly studied mechanism of intercellular communication that involves the transfer of molecules, including but not limited to proteins, lipids, and non-coding RNAs, to induce functional changes in the recipient cells. In this present mini-review, we focus on the PTM-regulated protein and non-coding RNA selection into eukaryotic EVs.
Collapse
Affiliation(s)
- Jonathan M Carnino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, United States
| | - Kareemah Ni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, United States
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
227
|
Mammalian CBX7 isoforms p36 and p22 exhibit differential responses to serum, varying functions for proliferation, and distinct subcellular localization. Sci Rep 2020; 10:8061. [PMID: 32415167 PMCID: PMC7228926 DOI: 10.1038/s41598-020-64908-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/20/2020] [Indexed: 01/04/2023] Open
Abstract
CBX7 is a polycomb group protein, and despite being implicated in many diseases, its role in cell proliferation has been controversial: some groups described its pro-proliferative properties, but others illustrated its inhibitory effects on cell growth. To date, the reason for the divergent observations remains unknown. While several isoforms for CBX7 were reported, no studies investigated whether the divergent roles of CBX7 could be due to distinct functions of CBX7 isoforms. In this study, we newly identified mouse CBX7 transcript variant 1 (mCbx7v1), which is homologous to the human CBX7 gene (hCBX7v1) and is expressed in various mouse organs. We revealed that mCbx7v1 and hCBX7v1 encode a 36 kDa protein (p36CBX7) whereas mCbx7 and hCBX7v3 encode a 22 kDa protein (p22CBX7). This study further demonstrated that p36CBX7 was localized to the nucleus and endogenously expressed in proliferating cells whereas p22CBX7 was localized to the cytoplasm, induced by serum starvation in both human and mouse cells, and inhibited cell proliferation. Together, these data indicate that CBX7 isoforms are localized in different locations in a cell and play differing roles in cell proliferation. This varying function of CBX7 isoforms may help us understand the distinct function of CBX7 in various studies.
Collapse
|
228
|
Celen AB, Sahin U. Sumoylation on its 25th anniversary: mechanisms, pathology, and emerging concepts. FEBS J 2020; 287:3110-3140. [DOI: 10.1111/febs.15319] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/04/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Arda B. Celen
- Department of Molecular Biology and Genetics Center for Life Sciences and Technologies Bogazici University Istanbul Turkey
| | - Umut Sahin
- Department of Molecular Biology and Genetics Center for Life Sciences and Technologies Bogazici University Istanbul Turkey
| |
Collapse
|
229
|
Cuomo O, Casamassa A, Brancaccio P, Laudati G, Valsecchi V, Anzilotti S, Vinciguerra A, Pignataro G, Annunziato L. Sumoylation of sodium/calcium exchanger in brain ischemia and ischemic preconditioning. Cell Calcium 2020; 87:102195. [DOI: 10.1016/j.ceca.2020.102195] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/10/2020] [Accepted: 03/13/2020] [Indexed: 11/26/2022]
|
230
|
Xu H, Wang H, Zhao W, Fu S, Li Y, Ni W, Xin Y, Li W, Yang C, Bai Y, Zhan M, Lu L. SUMO1 modification of methyltransferase-like 3 promotes tumor progression via regulating Snail mRNA homeostasis in hepatocellular carcinoma. Theranostics 2020; 10:5671-5686. [PMID: 32483411 PMCID: PMC7254988 DOI: 10.7150/thno.42539] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/13/2020] [Indexed: 01/03/2023] Open
Abstract
Rationale: Hepatocellular carcinoma (HCC) is one of the leading causes of mortality worldwide. Methyltransferase-like 3 (Mettl3), an RNA N6-methyladenosine (m6A) methyltransferase, has been shown to act as an oncogene in several human cancers. However, the regulatory role of posttranslational modifications of Mettl3 in liver cancer remains elusive. Methods: SUMOylation was analyzed using immunoprecipitation and western blot assays. In vitro and in vivo biological functions were examined using MTS, colony formation, wound healing, transwell, apoptosis, and viability assays and the BALB/c nude mouse model, respectively. Immunohistochemistry was conducted to evaluate the prognostic value of Mettl3 expression in HCC. The regulatory mechanism of Mettl3 in HCC was investigated by m6A dot blot, immunofluorescence, dual luciferase reporter, protein stability, and RNA stability assays. Results: Mettl3 was found to be SUMOylated by a small ubiquitin-like modifier SUMO1. Further, SUMOylation of Mettl3 was increased upon mitogen stimulation, which correlated with UBC9 upregulation, and was positively correlated with high metastatic potential of liver cancer. Finally, SUMOylation of Mettl3 was found to regulate HCC progression via controlling Snail mRNA homeostasis in an m6A methyltransferase activity-dependent manner. Conclusions: This study revealed a novel mechanism of SUMOylated Mettl3-mediated Snail mRNA homeostasis, identifying the UBC9/SUMOylated Mettl3/Snail axis as a novel mediator of the SUMO pathway involved in HCC progression.
Collapse
Affiliation(s)
- Hongfa Xu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Hao Wang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Wei Zhao
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Sirui Fu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Yong Li
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Wenjun Ni
- Department of urinary surgery, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Yongjie Xin
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Wei Li
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Chenzi Yang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Yanyan Bai
- School of Stomatology, Changsha Medical University, Changsha City, Hunan, 410219, China
| | - Meixiao Zhan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong, 519000, China
| |
Collapse
|
231
|
Petushkova AI, Zamyatnin AA. Redox-Mediated Post-Translational Modifications of Proteolytic Enzymes and Their Role in Protease Functioning. Biomolecules 2020; 10:biom10040650. [PMID: 32340246 PMCID: PMC7226053 DOI: 10.3390/biom10040650] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/13/2022] Open
Abstract
Proteolytic enzymes play a crucial role in metabolic processes, providing the cell with amino acids through the hydrolysis of multiple endogenous and exogenous proteins. In addition to this function, proteases are involved in numerous protein cascades to maintain cellular and extracellular homeostasis. The redox regulation of proteolysis provides a flexible dose-dependent mechanism for proteolytic activity control. The excessive reactive oxygen species (ROS) and reactive nitrogen species (RNS) in living organisms indicate pathological conditions, so redox-sensitive proteases can swiftly induce pro-survival responses or regulated cell death (RCD). At the same time, severe protein oxidation can lead to the dysregulation of proteolysis, which induces either protein aggregation or superfluous protein hydrolysis. Therefore, oxidative stress contributes to the onset of age-related dysfunction. In the present review, we consider the post-translational modifications (PTMs) of proteolytic enzymes and their impact on homeostasis.
Collapse
Affiliation(s)
- Anastasiia I. Petushkova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
232
|
Molecular mechanisms in SUMO conjugation. Biochem Soc Trans 2020; 48:123-135. [PMID: 31872228 DOI: 10.1042/bst20190357] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 01/25/2023]
Abstract
The small ubiquitin-like modifier (SUMO) is a post-translational modifier that can regulate the function of hundreds of proteins inside the cell. SUMO belongs to the ubiquitin-like family of proteins that can be attached to target proteins by a dedicated enzymatic cascade pathway formed by E1, E2 and E3 enzymes. SUMOylation is involved in many cellular pathways, having in most instances essential roles for their correct function. In this review, we want to highlight the latest research on the molecular mechanisms that lead to the formation of the isopeptidic bond between the lysine substrate and the C-terminus of SUMO. In particular, we will focus on the recent discoveries on the catalytic function of the SUMO E3 ligases revealed by structural and biochemical approaches. Also, we will discuss important questions regarding specificity in SUMO conjugation, which it still remains as a major issue due to the small number of SUMO E3 ligases discovered so far, in contrast with the large number of SUMO conjugated proteins in the cell.
Collapse
|
233
|
Wu R, Fang J, Liu M, A J, Liu J, Chen W, Li J, Ma G, Zhang Z, Zhang B, Fu L, Dong JT. SUMOylation of the transcription factor ZFHX3 at Lys-2806 requires SAE1, UBC9, and PIAS2 and enhances its stability and function in cell proliferation. J Biol Chem 2020; 295:6741-6753. [PMID: 32249212 DOI: 10.1074/jbc.ra119.012338] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/01/2020] [Indexed: 01/07/2023] Open
Abstract
SUMOylation is a posttranslational modification (PTM) at a lysine residue and is crucial for the proper functions of many proteins, particularly of transcription factors, in various biological processes. Zinc finger homeobox 3 (ZFHX3), also known as AT motif-binding factor 1 (ATBF1), is a large transcription factor that is active in multiple pathological processes, including atrial fibrillation and carcinogenesis, and in circadian regulation and development. We have previously demonstrated that ZFHX3 is SUMOylated at three or more lysine residues. Here, we investigated which enzymes regulate ZFHX3 SUMOylation and whether SUMOylation modulates ZFHX3 stability and function. We found that SUMO1, SUMO2, and SUMO3 each are conjugated to ZFHX3. Multiple lysine residues in ZFHX3 were SUMOylated, but Lys-2806 was the major SUMOylation site, and we also found that it is highly conserved among ZFHX3 orthologs from different animal species. Using molecular analyses, we identified the enzymes that mediate ZFHX3 SUMOylation; these included SUMO1-activating enzyme subunit 1 (SAE1), an E1-activating enzyme; SUMO-conjugating enzyme UBC9 (UBC9), an E2-conjugating enzyme; and protein inhibitor of activated STAT2 (PIAS2), an E3 ligase. Multiple analyses established that both SUMO-specific peptidase 1 (SENP1) and SENP2 deSUMOylate ZFHX3. SUMOylation at Lys-2806 enhanced ZFHX3 stability by interfering with its ubiquitination and proteasomal degradation. Functionally, Lys-2806 SUMOylation enabled ZFHX3-mediated cell proliferation and xenograft tumor growth of the MDA-MB-231 breast cancer cell line. These findings reveal the enzymes involved in, and the functional consequences of, ZFHX3 SUMOylation, insights that may help shed light on ZFHX3's roles in various cellular and pathophysiological processes.
Collapse
Affiliation(s)
- Rui Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jiali Fang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Mingcheng Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jun A
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jinming Liu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wenxuan Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Juan Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Gui Ma
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhiqian Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Baotong Zhang
- Emory Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jin-Tang Dong
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
234
|
Role of PML SUMOylation in arsenic trioxide-induced fibrosis in HSCs. Life Sci 2020; 251:117607. [PMID: 32240679 DOI: 10.1016/j.lfs.2020.117607] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/15/2020] [Accepted: 03/22/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Arsenic trioxide (ATO) can bind directly to the human promyelocytic leukemia (PML) protein, leading to modification of PML by SUMOs. UBC9 is the only known E2-conjugating enzyme involved in SUMOylation. PML degradation via RNF4, an E3 ubiquitin ligases family member. PML is key organizer of nuclear bodies (NBs) that regulate many biological processes such as senescence, and DNA damage. ATO can activate the TGFβ/Smad signaling pathway, causing liver fibrosis. However, the roles of PML Sumoylation in ATO-induced liver fibrosis remain unclear. OBJECTIVE This study aimed to investigate the role of PML Sumoylation in the ATO-induced HSCs activation and to improve the mechanism of ATO-induced liver fibrosis. METHODS Hepatic stellate cells (HSCs) were treated with 2 μmol/L ATO. Cell viability was detected by CCK-8 analysis. Immunoblot analysis and real-time quantitative PCR were used to detect the expression of IL-1β, TNF-α, TGF-β1, p-Smad2/3, α-SMA, Collagen I and PML SUMOylation after silencing PML, UBC9, and RNF4, respectively. The formation of PML-NBs was observed by immunofluorescence staining. RESULTS 2 and 5 μmol/L ATO intervention increased HSCs cell viability. ATO was able to significantly trigger PML SUMOylation and the formation of PML-NBs. Inhibition of SUMOylated PML by silencing UBC9, subsequently preventing the downregulation of HSCs activation indicators induced by ATO (P < 0.05). Conversely, enhancing SUMOylated PML accumulation by silencing RNF4, activating TGFβ/Smad signaling pathway, eventually promoting the induction of liver fibrosis. CONCLUSION These results indicated that PML SUMOylation plays a critical role in the development of liver fibrosis induced by ATO.
Collapse
|
235
|
Zárraga-Granados G, Muciño-Hernández G, Sánchez-Carbente MR, Villamizar-Gálvez W, Peñas-Rincón A, Arredondo C, Andrés ME, Wood C, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 is regulated by SUMO modification to induce autophagic cell death. PLoS One 2020; 15:e0222072. [PMID: 32210435 PMCID: PMC7094859 DOI: 10.1371/journal.pone.0222072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/08/2020] [Indexed: 12/31/2022] Open
Abstract
NR4A is a nuclear receptor protein family whose members act as sensors of cellular environment and regulate multiple processes such as metabolism, proliferation, migration, apoptosis, and autophagy. Since the ligand binding domains of these receptors have no cavity for ligand interaction, their function is most likely regulated by protein abundance and post-translational modifications. In particular, NR4A1 is regulated by protein abundance, phosphorylation, and subcellular distribution (nuclear-cytoplasmic translocation), and acts both as a transcription factor and as a regulator of other interacting proteins. SUMOylation is a post-translational modification that can affect protein stability, transcriptional activity, alter protein-protein interactions and modify intracellular localization of target proteins. In the present study we evaluated the role of SUMOylation as a posttranslational modification that can regulate the activity of NR4A1 to induce autophagy-dependent cell death. We focused on a model potentially relevant for neuronal cell death and demonstrated that NR4A1 needs to be SUMOylated to induce autophagic cell death. We observed that a triple mutant in SUMOylation sites has reduced SUMOylation, increased transcriptional activity, altered intracellular distribution, and more importantly, its ability to induce autophagic cell death is impaired.
Collapse
Affiliation(s)
- Gabriela Zárraga-Granados
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Gabriel Muciño-Hernández
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - María R. Sánchez-Carbente
- Biotechnology Research Center, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Wendy Villamizar-Gálvez
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Ana Peñas-Rincón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Cristian Arredondo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María E. Andrés
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christopher Wood
- Laboratorio Nacional de Microscopía Avanzada, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Luis Covarrubias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Susana Castro-Obregón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
- * E-mail:
| |
Collapse
|
236
|
Xu Y, Wu W, Han Q, Wang Y, Li C, Zhang P, Xu H. Post-translational modification control of RNA-binding protein hnRNPK function. Open Biol 2020; 9:180239. [PMID: 30836866 PMCID: PMC6451366 DOI: 10.1098/rsob.180239] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Heterogeneous nuclear ribonucleoprotein K (hnRNPK), a ubiquitously occurring RNA-binding protein (RBP), can interact with numerous nucleic acids and various proteins and is involved in a number of cellular functions including transcription, translation, splicing, chromatin remodelling, etc. Through its abundant biological functions, hnRNPK has been implicated in cellular events including proliferation, differentiation, apoptosis, DNA damage repair and the stress and immune responses. Thus, it is critical to understand the mechanism of hnRNPK regulation and its downstream effects on cancer and other diseases. A number of recent studies have highlighted that several post-translational modifications (PTMs) possibly play an important role in modulating hnRNPK function. Phosphorylation is the most widely occurring PTM in hnRNPK. For example, in vivo analyses of sites such as S116 and S284 illustrate the purpose of PTM of hnRNPK in altering its subcellular localization and its ability to bind target nucleic acids or proteins. Other PTMs such as methylation, ubiquitination, sumoylation, glycosylation and proteolytic cleavage are increasingly implicated in the regulation of DNA repair, cellular stresses and tumour growth. In this review, we describe the PTMs that impact upon hnRNPK function on gene expression programmes and different disease states. This knowledge is key in allowing us to better understand the mechanism of hnRNPK regulation.
Collapse
Affiliation(s)
- Yongjie Xu
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| | - Wei Wu
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| | - Qiu Han
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| | - Yaling Wang
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| | - Cencen Li
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| | - Pengpeng Zhang
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| | - Haixia Xu
- College of Life Science, Xinyang Normal University , Xinyang 464000 , People's Republic of China
| |
Collapse
|
237
|
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
|
238
|
Rabellino A, Khanna KK. The implication of the SUMOylation pathway in breast cancer pathogenesis and treatment. Crit Rev Biochem Mol Biol 2020; 55:54-70. [PMID: 32183544 DOI: 10.1080/10409238.2020.1738332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most commonly diagnosed malignancy in woman worldwide, and is the second most common cause of death in developed countries. The transformation of a normal cell into a malignant derivate requires the acquisition of diverse genomic and proteomic changes, including enzymatic post-translational modifications (PTMs) on key proteins encompassing critical cell signaling events. PTMs occur on proteins after translation, and regulate several aspects of proteins activity, including their localization, activation and turnover. Deregulation of PTMs can potentially lead to tumorigenesis, and several de-regulated PTM pathways contribute to abnormal cell proliferation during breast tumorigenesis. SUMOylation is a PTM that plays a pivotal role in numerous aspects of cell physiology, including cell cycle regulation, protein trafficking and turnover, and DNA damage repair. Consistently with this, the deregulation of the SUMO pathway is observed in different human pathologies, including breast cancer. In this review we will describe the role of SUMOylation in breast tumorigenesis and its implication for breast cancer therapy.
Collapse
Affiliation(s)
- Andrea Rabellino
- QIMR Berghofer Medical Research Institute, Brisbane City, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane City, Australia
| |
Collapse
|
239
|
Viral DNA Binding Protein SUMOylation Promotes PML Nuclear Body Localization Next to Viral Replication Centers. mBio 2020; 11:mBio.00049-20. [PMID: 32184235 PMCID: PMC7078464 DOI: 10.1128/mbio.00049-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human adenoviruses (HAdVs) have developed mechanisms to manipulate cellular antiviral measures to ensure proper DNA replication, with detailed processes far from being understood. Host cells repress incoming viral genomes through a network of transcriptional regulators that normally control cellular homeostasis. The nuclear domains involved are promyelocytic leukemia protein nuclear bodies (PML-NBs), interferon-inducible, dot-like nuclear structures and hot spots of SUMO posttranslational modification (PTM). In HAdV-infected cells, such SUMO factories are found in close proximity to newly established viral replication centers (RCs) marked by the adenoviral DNA binding protein (DBP) E2A. Here, we show that E2A is a novel target of host SUMOylation, leading to PTMs supporting E2A function in promoting productive infection. Our data show that SUMOylated E2A interacts with PML. Decreasing SUMO-E2A protein levels by generating HAdV variants mutated in the three main SUMO conjugation motifs (SCMs) led to lower numbers of viral RCs and PML-NBs, and these two structures were no longer next to each other. Our data further indicate that SUMOylated E2A binds the host transcription factor Sp100A, promoting HAdV gene expression, and represents the molecular bridge between PML tracks and adjacent viral RCs. Consequently, E2A SCM mutations repressed late viral gene expression and progeny production. These data highlight a novel mechanism used by the virus to benefit from host antiviral responses by exploiting the cellular SUMO conjugation machinery.IMPORTANCE PML nuclear bodies (PML-NBs) are implicated in general antiviral defense based on recruiting host restriction factors; however, it is not understood so far why viruses would establish viral replication centers (RCs) juxtaposed to such "antiviral" compartments. To understand this enigma, we investigate the cross talk between PML-NB components and viral RCs to find the missing link connecting both compartments to promote efficient viral replication and gene expression. Taken together, the current concept is more intricate than originally believed, since viruses apparently take advantage of several specific PML-NB-associated proteins to promote productive infection. Simultaneously, they efficiently inhibit antiviral measures to maintain the viral infectious program. Our data provide evidence that SUMOylation of the viral RC marker protein E2A represents the basis of this virus-host interface and regulates various downstream events to support HAdV productive infection. These results are the basis of our current attempts to generate and screen for specific E2A SUMOylation inhibitors to constitute novel therapeutic approaches to limit and prevent HAdV-mediated diseases and mortality of immunosuppressed patients.
Collapse
|
240
|
RNF4-mediated SUMO-targeted ubiquitination relieves PARIS/ZNF746-mediated transcriptional repression. Biochem Biophys Res Commun 2020; 526:110-116. [PMID: 32197837 DOI: 10.1016/j.bbrc.2020.03.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/10/2020] [Indexed: 01/28/2023]
Abstract
The transcriptional repressor PARIS, which is a substrate of the ubiquitin E3 ligase parkin, represses the expression of the transcriptional co-activator, PGC-1α. However, little is known about how its repression activity is regulated. We have previously shown that PARIS is SUMOylated, and this SUMOylation plays an important role in regulating its transcriptional repression activity. In this study, we demonstrated that PARIS SUMOylation induced its ubiquitination and subsequent proteasomal degradation, which was mediated by the SUMO-targeted ubiquitin ligase RNF4. Reporter gene assays revealed that co-expression of SUMO3 and RNF4 relieved PARIS-mediated transcriptional repression. Conversely, the SUMO E3 ligase PIASy inhibited the RNF4-mediated ubiquitination of PARIS and blocked the RNF4-mediated relief of PARIS-mediated transcriptional repression. These results suggest that RNF4 regulates PARIS ubiquitination to control its transcriptional repression activity.
Collapse
|
241
|
Le-Trilling VTK, Trilling M. Ub to no good: How cytomegaloviruses exploit the ubiquitin proteasome system. Virus Res 2020; 281:197938. [PMID: 32198076 DOI: 10.1016/j.virusres.2020.197938] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/17/2022]
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous member of the Betaherpesvirinae subfamily, causing life-threatening diseases in individuals with impaired, immature, or senescent immunity. Accordingly, HIV-infected AIDS patients, transplant recipients, and congenitally infected neonates frequently suffer from symptomatic episodes of HCMV replication. Like all viruses, HCMV has a split relationship with the host proteome. Efficient virus replication can only be achieved if proteins involved in intrinsic, innate, and adaptive immune responses are sufficiently antagonized. Simultaneously, the abundance and function of proteins involved in the synthesis of chemical building blocks required for virus production, such as nucleotides, amino acids, and fatty acids, must be preserved or even enriched. The ubiquitin (Ub) proteasome system (UPS) constitutes one of the most relevant protein decay systems of eukaryotic cells. In addition to the regulation of the turn-over and abundance of thousands of proteins, the UPS also generates the majority of peptides presented by major histocompatibility complex (MHC) molecules to allow surveillance by T lymphocytes. Cytomegaloviruses exploit the UPS to regulate the abundance of viral proteins and to manipulate the host proteome in favour of viral replication and immune evasion. After summarizing the current knowledge of CMV-mediated misuse of the UPS, we discuss the evolution of viral proteins utilizing the UPS for the degradation of defined target proteins. We propose two alternative routes of adapter protein development and their mechanistic consequences.
Collapse
Affiliation(s)
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
242
|
Sahu MS, Patra S, Kumar K, Kaur R. SUMOylation in Human Pathogenic Fungi: Role in Physiology and Virulence. J Fungi (Basel) 2020; 6:E32. [PMID: 32143470 PMCID: PMC7096222 DOI: 10.3390/jof6010032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023] Open
Abstract
The small ubiquitin-related modifier (SUMO) protein is an important component of the post-translational protein modification systems in eukaryotic cells. It is known to modify hundreds of proteins involved in diverse cellular processes, ranging from nuclear pore dynamics to signal transduction pathways. Owing to its reversible nature, the SUMO-conjugation of proteins (SUMOylation) holds a prominent place among mechanisms that regulate the functions of a wide array of cellular proteins. The dysfunctional SUMOylation system has been associated with many human diseases, including neurodegenerative and autoimmune disorders. Furthermore, the non-pathogenic yeast Saccharomyces cerevisiae has served as an excellent model to advance our understanding of enzymes involved in SUMOylation and proteins modified by SUMOylation. Taking advantage of the tools and knowledge obtained from the S. cerevisiae SUMOylation system, research on fungal SUMOylation is beginning to gather pace, and new insights into the role of SUMOylation in the pathobiology of medically important fungi are emerging. Here, we summarize the known information on components of the SUMOylation machinery, and consequences of overexpression or deletion of these components in the human pathogenic fungi, with major focus on two prevalent Candida bloodstream pathogens, C. albicans and C. glabrata. Additionally, we have identified SUMOylation components, through in silico analysis, in four medically relevant fungi, and compared their sequence similarity with S. cerevisiae counterparts. SUMOylation modulates the virulence of C. albicans and C. glabrata, while it is required for conidia production in Aspergillus nidulans and A. flavus. In addition to highlighting these recent developments, we discuss how SUMOylation fine tunes the expression of virulence factors, and influences survival of fungal cells under diverse stresses in vitro and in the mammalian host.
Collapse
Affiliation(s)
- Mahima Sagar Sahu
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, Telangana, India; (M.S.S.); (S.P.); (K.K.)
- Graduate studies, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Sandip Patra
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, Telangana, India; (M.S.S.); (S.P.); (K.K.)
- Graduate studies, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Kundan Kumar
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, Telangana, India; (M.S.S.); (S.P.); (K.K.)
- Graduate studies, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Rupinder Kaur
- Laboratory of Fungal Pathogenesis, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, Telangana, India; (M.S.S.); (S.P.); (K.K.)
| |
Collapse
|
243
|
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
|
244
|
Lotz C, Lamour V. The interplay between DNA topoisomerase 2α post-translational modifications and drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:149-160. [PMID: 35582608 PMCID: PMC9090595 DOI: 10.20517/cdr.2019.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/19/2020] [Accepted: 02/05/2020] [Indexed: 01/13/2023]
Abstract
The type 2 DNA topoisomerases (Top2) are conserved enzymes and biomarkers for cell proliferation. The catalytic activities of the human isoform Top2α are essential for the regulation of DNA topology during DNA replication, transcription, and chromosome segregation. Top2α is a prominent target for anti-cancer drugs and is highly regulated by post-translational modifications (PTM). Despite an increasing number of proteomic studies, the extent of PTM in cancer cells and its importance in drug response remains largely uncharacterized. In this review, we highlight the different modifications affecting the human Top2α in healthy and cancer cells, taking advantage of the structure-function information accumulated in the past decades. We also overview the regulation of Top2α by PTM, the level of PTM in cancer cells, and the resistance to therapeutic compounds targeting the Top2 enzyme. Altogether, this review underlines the importance of future studies addressing more systematically the interplay between PTM and Top2 drug resistance.
Collapse
Affiliation(s)
- Christophe Lotz
- Integrative Structural Biology Department, IGBMC, Université de Strasbourg, CNRS UMR 7104, INSERM U1258, Illkirch 67404, France
| | - Valérie Lamour
- Integrative Structural Biology Department, IGBMC, Université de Strasbourg, CNRS UMR 7104, INSERM U1258, Illkirch 67404, France
- Hôpitaux Universitaires de Strasbourg, Strasbourg 67000, France
| |
Collapse
|
245
|
Poreba E, Durzynska J. Nuclear localization and actions of the insulin-like growth factor 1 (IGF-1) system components: Transcriptional regulation and DNA damage response. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 784:108307. [PMID: 32430099 DOI: 10.1016/j.mrrev.2020.108307] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor (IGF) system stimulates growth, proliferation, and regulates differentiation of cells in a tissue-specific manner. It is composed of two insulin-like growth factors (IGF-1 and IGF-2), six insulin-like growth factor-binding proteins (IGFBPs), and two insulin-like growth factor receptors (IGF-1R and IGF-2R). IGF actions take place mostly through the activation of the plasma membrane-bound IGF-Rs by the circulating ligands (IGFs) released from the IGFBPs that stabilize their levels in the serum. This review focuses on the IGF-1 part of the system. The IGF-1 gene, which is expressed mainly in the liver as well as in other tissues, comprises six alternatively spliced exons that code for three protein isoforms (pro-IGF-1A, pro-IGF-1B, and pro-IGF-1C), which are processed to mature IGF-1 and E-peptides. The IGF-1R undergoes autophosphorylation, resulting in a signaling cascade involving numerous cytoplasmic proteins such as AKT and MAPKs, which regulate the expression of target genes. However, a more complex picture of the axis has recently emerged with all its components being translocated to the nuclear compartment. IGF-1R takes part in the regulation of gene expression by forming transcription complexes, modifying the activity of chromatin remodeling proteins, and participating in DNA damage tolerance mechanisms. Four IGFBPs contain a nuclear localization signal (NLS), which targets them to the nucleus, where they regulate gene expression (IGFBP-2, IGFBP-3, IGFBP-5, IGFBP-6) and DNA damage repair (IGFBP-3 and IGFBP-6). Last but not least, the IGF-1B isoform has been reported to be localized in the nuclear compartment. However, no specific molecular actions have been assigned to the nuclear pro-IGF-1B or its derivative EB peptide. Therefore, further studies are needed to shed light on their nuclear activity. These recently uncovered nuclear actions of different components of the IGF-1 axis are relevant in cancer cell biology and are discussed in this review.
Collapse
Affiliation(s)
- Elzbieta Poreba
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| | - Julia Durzynska
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
246
|
Rosário-Ferreira N, Preto AJ, Melo R, Moreira IS, Brito RMM. The Central Role of Non-Structural Protein 1 (NS1) in Influenza Biology and Infection. Int J Mol Sci 2020; 21:E1511. [PMID: 32098424 PMCID: PMC7073157 DOI: 10.3390/ijms21041511] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 01/07/2023] Open
Abstract
Influenza (flu) is a contagious viral disease, which targets the human respiratory tract and spreads throughout the world each year. Every year, influenza infects around 10% of the world population and between 290,000 and 650,000 people die from it according to the World Health Organization (WHO). Influenza viruses belong to the Orthomyxoviridae family and have a negative sense eight-segment single-stranded RNA genome that encodes 11 different proteins. The only control over influenza seasonal epidemic outbreaks around the world are vaccines, annually updated according to viral strains in circulation, but, because of high rates of mutation and recurrent genetic assortment, new viral strains of influenza are constantly emerging, increasing the likelihood of pandemics. Vaccination effectiveness is limited, calling for new preventive and therapeutic approaches and a better understanding of the virus-host interactions. In particular, grasping the role of influenza non-structural protein 1 (NS1) and related known interactions in the host cell is pivotal to better understand the mechanisms of virus infection and replication, and thus propose more effective antiviral approaches. In this review, we assess the structure of NS1, its dynamics, and multiple functions and interactions, to highlight the central role of this protein in viral biology and its potential use as an effective therapeutic target to tackle seasonal and pandemic influenza.
Collapse
Affiliation(s)
- Nícia Rosário-Ferreira
- Coimbra Chemistry Center, Chemistry Department, Faculty of Science and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology. University of Coimbra, UC Biotech Building, 3060-197 Cantanhede, Portugal
| | - António J. Preto
- CNC—Center for Neuroscience and Cell Biology. University of Coimbra, UC Biotech Building, 3060-197 Cantanhede, Portugal
| | - Rita Melo
- CNC—Center for Neuroscience and Cell Biology. University of Coimbra, UC Biotech Building, 3060-197 Cantanhede, Portugal
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| | - Irina S. Moreira
- CNC—Center for Neuroscience and Cell Biology. University of Coimbra, UC Biotech Building, 3060-197 Cantanhede, Portugal
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Rui M. M. Brito
- Coimbra Chemistry Center, Chemistry Department, Faculty of Science and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
247
|
Brix DM, Bundgaard Clemmensen KK, Kallunki T. Zinc Finger Transcription Factor MZF1-A Specific Regulator of Cancer Invasion. Cells 2020; 9:cells9010223. [PMID: 31963147 PMCID: PMC7016646 DOI: 10.3390/cells9010223] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/11/2022] Open
Abstract
Over 90% of cancer deaths are due to cancer cells metastasizing into other organs. Invasion is a prerequisite for metastasis formation. Thus, inhibition of invasion can be an efficient way to prevent disease progression in these patients. This could be achieved by targeting the molecules regulating invasion. One of these is an oncogenic transcription factor, Myeloid Zinc Finger 1 (MZF1). Dysregulated transcription factors represent a unique, increasing group of drug targets that are responsible for aberrant gene expression in cancer and are important nodes driving cancer malignancy. Recent studies report of a central involvement of MZF1 in the invasion and metastasis of various solid cancers. In this review, we summarize the research on MZF1 in cancer including its function and role in lysosome-mediated invasion and in the expression of genes involved in epithelial to mesenchymal transition. We also discuss possible means to target it on the basis of the current knowledge of its function in cancer.
Collapse
Affiliation(s)
- Ditte Marie Brix
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; (D.M.B.); (K.K.B.C.)
- Danish Medicines Council, Dampfærgevej 27-29, 2100 Copenhagen, Denmark
| | - Knut Kristoffer Bundgaard Clemmensen
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; (D.M.B.); (K.K.B.C.)
| | - Tuula Kallunki
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; (D.M.B.); (K.K.B.C.)
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence: ; Tel.: +45-35-25-7746
| |
Collapse
|
248
|
Du CP, Wang M, Geng C, Hu B, Meng L, Xu Y, Cheng B, Wang N, Zhu QJ, Hou XY. Activity-Induced SUMOylation of Neuronal Nitric Oxide Synthase Is Associated with Plasticity of Synaptic Transmission and Extracellular Signal-Regulated Kinase 1/2 Signaling. Antioxid Redox Signal 2020; 32:18-34. [PMID: 31642335 DOI: 10.1089/ars.2018.7669] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Aims: Neuronal nitric oxide synthase (nNOS) and nitric oxide (NO) signaling have been implicated in learning, memory, and underlying long-lasting synaptic plasticity. In this study, we aimed at detecting whether nNOS is a target protein of SUMOylation in the hippocampus and its contributions to hippocampal long-term potentiation (LTP) of synaptic transmission. Results: We showed that N-methyl-d-aspartate receptor-dependent neuronal activity enhancement induced the attachment of small ubiquitin-like modifier 1 (SUMO1) to nNOS. Protein inhibitor of activated STAT3 (PIAS3) promoted SUMO1 conjugation at K725 and K739 on nNOS, which upregulated NO production and nNOS S1412 phosphorylation (activation). In addition, the N-terminus (amino acids 43-86) of PIAS3 bound nNOS directly. Tat-tagged PIAS3 segment representing amino acids 43-86, a cell-permeable peptide containing PIAS3 residues 43-86, suppressed activity-induced nNOS SUMOylation by disrupting PIAS3-nNOS association. It also decreased LTP-related expression of Arc and brain-derived neurotrophic factor and blocked signaling via extracellular signal-regulated kinase (ERK) 1/2 and Elk-1 in the hippocampus. More importantly, PIAS3-mediated nNOS SUMOylation was required for activity-regulated ERK1/2 activation in nNOS-positive neurons and hippocampal LTP induction. Innovation and Conclusion: These findings indicated that network activity-regulated nNOS SUMOylation underlies excitatory synaptic LTP by facilitating nNOS-NO-ERK1/2 signal cascades.
Collapse
Affiliation(s)
- Cai-Ping Du
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China.,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mei Wang
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Chi Geng
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China.,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bin Hu
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Li Meng
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Yan Xu
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Bao Cheng
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Nan Wang
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Qiu-Ju Zhu
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Yu Hou
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China.,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
249
|
Mohammed Y, Palmblad M. Using the Object-Oriented PowerShell for Simple Proteomics Data Analysis. Methods Mol Biol 2020; 2051:389-405. [PMID: 31552639 DOI: 10.1007/978-1-4939-9744-2_17] [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] [Indexed: 06/10/2023]
Abstract
Scripting languages such as Python and Bash are appreciated for solving simple, everyday tasks in bioinformatics. A more recent, object-oriented command shell and scripting language, PowerShell, has many attractive features: an object-oriented interactive command line, fluent navigation and manipulation of XML files, ability to explore and consume Web services from the command line, consistent syntax and grammar, rich regular expressions, and advanced output formatting. The key difference between classical command shells and scripting languages, such as bash, and object-oriented ones, such as PowerShell, is that in the latter the result of a command is a structured object with inherited properties and methods rather than a simple stream of characters. Conveniently, PowerShell is included in all new releases of Microsoft Windows and is available for Linux and macOS, making any data processing script portable. In this chapter we demonstrate how PowerShell in particular allows easy interaction with mass spectrometry data in XML formats, connection to Web services for tools such as BLAST, and presentation of results as formatted text or graphics. These features make PowerShell much more than "yet another scripting language."
Collapse
Affiliation(s)
- Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria, BC, Canada.
| | - Magnus Palmblad
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
250
|
Liberman AC, Budziñski ML, Sokn C, Gobbini RP, Ugo MB, Arzt E. SUMO conjugation as regulator of the glucocorticoid receptor-FKBP51 cellular response to stress. Steroids 2020; 153:108520. [PMID: 31604074 DOI: 10.1016/j.steroids.2019.108520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 01/19/2023]
Abstract
In order to adequately respond to stressful stimuli, glucocorticoids (GCs) target almost every tissue of the body. By exerting a negative feedback loop in the hypothalamic-pituitary-adrenal (HPA) axis GCs inhibit their own synthesis and restore homeostasis. GCs actions are mostly mediated by the GC receptor (GR), a member of the nuclear receptor superfamily. Alterations of the GR activity have been associatedto different diseases including mood disorders and can lead to severe complication. Therefore, understanding the molecular complexity of GR modulation is mandatory for the development of new and effective drugs for treating GR-associated disorders. FKBP51 is a GR chaperone that has gained much attention because it is a strong inhibitor of GR activity and has a crucial role in psychiatric diseases. Both GR and FKBP51 activity are regulated by SUMOylation, a posttranslational (PTM). In this review, we focus on the impact of SUMO-conjugation as a regulator of this pathway.
Collapse
Affiliation(s)
- Ana C Liberman
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina.
| | - Maia L Budziñski
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Clara Sokn
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Romina P Gobbini
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Maria B Ugo
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)- CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina; Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
| |
Collapse
|