51
|
Wada H, Suzuki D, Niikura T. Regulation of ALS-Associated SOD1 Mutant SUMOylation and Aggregation by SENP and PIAS Family Proteins. J Mol Neurosci 2020; 70:2007-2014. [PMID: 32462635 DOI: 10.1007/s12031-020-01604-w] [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: 12/15/2019] [Accepted: 05/19/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease specific to motor neurons. Pathogenic mutations in an ALS-associated gene encoding superoxide dismutase 1 (SOD1) have been identified in familial ALS (fALS) cases. SOD1 with fALS-linked mutations is prone to form cytotoxic aggregates that cause cellular dysfunction. We previously demonstrated that the modification of SOD1 by small ubiquitin-like modifier (SUMO) 3 enhances the aggregation of fALS-linked SOD1 mutants. SUMOylation is a reversible post-translational modification targeting lysine residues. SUMO conjugation is mediated by the enzymes E1, E2, and E3, and deconjugation is catalyzed by deSUMOylation enzymes. To understand the process of SOD1 aggregation, we examined the involvement of protein inhibitor of activated STAT (PIAS) family and sentrin-specific protease (SENP) family proteins in the SUMOylation of SOD1 mutants. We found that all four types of PIAS family proteins, E3 ligase of SUMOylation, increased SUMOylation of SOD1 mutants. Among three SENP family proteins tested, deSUMOylation enzymes, SENP1, exhibited the most efficient deconjugation effect. In co-expression experiments, PIASy and SENP1 increased and decreased the number of cells exhibiting SOD1-mutant aggregation, respectively, confirming the effect of these enzymes on SOD1 aggregation. These findings suggest that regulation of SUMOylation affects the pathogenesis of ALS.
Collapse
Affiliation(s)
- Harmony Wada
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo, 102-8554, Japan
| | - Dan Suzuki
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo, 102-8554, Japan
| | - Takako Niikura
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo, 102-8554, Japan.
| |
Collapse
|
52
|
Liu Z, Tardat M, Gill ME, Royo H, Thierry R, Ozonov EA, Peters AH. SUMOylated PRC1 controls histone H3.3 deposition and genome integrity of embryonic heterochromatin. EMBO J 2020; 39:e103697. [PMID: 32395866 PMCID: PMC7327501 DOI: 10.15252/embj.2019103697] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Chromatin integrity is essential for cellular homeostasis. Polycomb group proteins modulate chromatin states and transcriptionally repress developmental genes to maintain cell identity. They also repress repetitive sequences such as major satellites and constitute an alternative state of pericentromeric constitutive heterochromatin at paternal chromosomes (pat‐PCH) in mouse pre‐implantation embryos. Remarkably, pat‐PCH contains the histone H3.3 variant, which is absent from canonical PCH at maternal chromosomes, which is marked by histone H3 lysine 9 trimethylation (H3K9me3), HP1, and ATRX proteins. Here, we show that SUMO2‐modified CBX2‐containing Polycomb Repressive Complex 1 (PRC1) recruits the H3.3‐specific chaperone DAXX to pat‐PCH, enabling H3.3 incorporation at these loci. Deficiency of Daxx or PRC1 components Ring1 and Rnf2 abrogates H3.3 incorporation, induces chromatin decompaction and breakage at PCH of exclusively paternal chromosomes, and causes their mis‐segregation. Complementation assays show that DAXX‐mediated H3.3 deposition is required for chromosome stability in early embryos. DAXX also regulates repression of PRC1 target genes during oogenesis and early embryogenesis. The study identifies a novel critical role for Polycomb in ensuring heterochromatin integrity and chromosome stability in mouse early development.
Collapse
Affiliation(s)
- Zichuan Liu
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Mathieu Tardat
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Mark E Gill
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Helene Royo
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Raphael Thierry
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Evgeniy A Ozonov
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Antoine Hfm Peters
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,Faculty of Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
53
|
Huang D, Liu H, Zhu A, Zhou Y, Li Y. Forebrain excitatory neuron-specific SENP2 knockout mouse displays hyperactivity, impaired learning and memory, and anxiolytic-like behavior. Mol Brain 2020; 13:59. [PMID: 32290845 PMCID: PMC7155287 DOI: 10.1186/s13041-020-00591-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
Sentrin/SUMO-specific protease 2 (SENP2) is a member of SENPs family involved in maturation of SUMO precursors and deSUMOylation of specific target, and is highly expressed in the central nervous system (CNS). Although SENP2 has been shown to modulate embryonic development, fatty acid metabolism, atherosclerosis and epilepsy, the function of SENP2 in the CNS remains poorly understood. To address the role of SENP2 in the CNS and its potential involvement in neuropathology, we generated SENP2 conditional knockout mice by crossing floxed SENP2 mice with CaMKIIα-Cre transgenic mice. Behavioral tests revealed that SENP2 ablation induced hyper-locomotor activity, anxiolytic-like behaviors, spatial working memory impairment and fear-associated learning defect. In line with these observations, our RNA sequencing (RNA-seq) data identified a variety of differential expression genes that are particularly enriched in locomotion, learning and memory related biologic process. Taken together, our results indicated that SENP2 plays a critical role in emotional and cognitive regulation. This SENP2 conditional knockout mice model may help reveal novel mechanisms that underlie a variety of neuropsychiatric disorders associated with anxiety and cognition.
Collapse
Affiliation(s)
- Dehua Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, 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, 280 South Chongqing Road, 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, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yi Zhou
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
54
|
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
|
55
|
Modification of cardiac transcription factor Gata6 by SUMO. Biochimie 2020; 170:212-218. [DOI: 10.1016/j.biochi.2020.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/31/2020] [Indexed: 01/25/2023]
|
56
|
Yu HI, Hsu T, Maruyama EO, Paschen W, Yang W, Hsu W. The requirement of SUMO2/3 for SENP2 mediated extraembryonic and embryonic development. Dev Dyn 2020; 249:237-244. [PMID: 31625212 PMCID: PMC7027852 DOI: 10.1002/dvdy.125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/23/2022] Open
Abstract
Small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is essential for the development of healthy placenta. The loss of SENP2 causes severe placental deficiencies and leads to embryonic death that is associated with heart and brain deformities. However, tissue-specific disruption of SENP2 demonstrates its dispensable role in embryogenesis and the embryonic defects are secondary to placental insufficiency. SENP2 regulates SUMO1 modification of Mdm2, which controls p53 activities critical for trophoblast cell proliferation and differentiation. Here we use genetic analyses to examine the involvement of SUMO2 and SUMO3 for SENP2-mediated placentation. The results indicate that hyper-SUMOylation caused by SENP2 deficiency can be compensated by reducing the level of SUMO modifiers. The placental deficiencies caused by the loss of SENP2 can be alleviated by the inactivation of gene encoding SUMO2 or SUMO3. Our findings demonstrate that SENP2 genetically interacts with SUMO2 and SUMO3 pivotal for the development of three major trophoblast layers. The alleviation of placental defects in the SENP2 knockouts further leads to the proper formation of the heart structures, including atrioventricular cushion and myocardium. SUMO2 and SUMO3 modifications regulate placentation and organogenesis mediated by SENP2.
Collapse
Affiliation(s)
- H‐M Ivy Yu
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
| | - Trunee Hsu
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
- Pittsford Mendon High SchoolPittsfordNew York
| | - Eri O Maruyama
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
| | - Wulf Paschen
- Department of AnesthesiologyDuke UniversityDurhamNorth Carolina
| | - Wei Yang
- Department of AnesthesiologyDuke UniversityDurhamNorth Carolina
| | - Wei Hsu
- Center for Oral BiologyUniversity of Rochester Medical CenterRochesterNew York
- Department of Biomedical GeneticsUniversity of Rochester Medical CenterRochesterNew York
- Stem Cell and Regenerative Medicine Institute, University of Rochester Medical CenterRochesterNew York
| |
Collapse
|
57
|
Kim YR, Jacobs JS, Li Q, Gaddam RR, Vikram A, Liu J, Kassan M, Irani K, Kumar S. SUMO2 regulates vascular endothelial function and oxidative stress in mice. Am J Physiol Heart Circ Physiol 2019; 317:H1292-H1300. [DOI: 10.1152/ajpheart.00530.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SUMOylation is a posttranslational modification of lysine residues. Modification of proteins by small ubiquitin-like modifiers (SUMO)1, -2, and -3 can achieve varied, and often unique, physiological and pathological effects. We looked for SUMO2-specific effects on vascular endothelial function. SUMO2 expression was upregulated in the aortic endothelium of hypercholesterolemic low-density lipoprotein receptor-deficient mice and was responsible for impairment of endothelium-dependent vasorelaxation in these mice. Moreover, overexpression of SUMO2 in aortas ex vivo, in cultured endothelial cells, and transgenically in the endothelium of mice increased vascular oxidative stress and impaired endothelium-dependent vasorelaxation. Conversely, inhibition of SUMO2 impaired physiological endothelium-dependent vasorelaxation in normocholesterolemic mice. These findings indicate that while endogenous SUMO2 is important in maintenance of normal endothelium-dependent vascular function, its upregulation impairs vascular homeostasis and contributes to hypercholesterolemia-induced endothelial dysfunction. NEW & NOTEWORTHY Sumoylation is known to impair vascular function; however, the role of specific SUMOs in the regulation of vascular function is not known. Using multiple complementary approaches, we show that hyper-SUMO2ylation impairs vascular endothelial function and increases vascular oxidative stress, whereas endogenous SUMO2 is essential for maintenance of normal physiological function of the vascular endothelium.
Collapse
Affiliation(s)
- Young-Rae Kim
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Julia S. Jacobs
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Qiuxia Li
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Ravinder Reddy Gaddam
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Ajit Vikram
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jing Liu
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Modar Kassan
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kaikobad Irani
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa City Veterans Affairs Medical Center, Iowa City, Iowa
| | - Santosh Kumar
- Department of Internal Medicine, Division of Cardiovascular Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
58
|
Oh JG, Watanabe S, Lee A, Gorski PA, Lee P, Jeong D, Liang L, Liang Y, Baccarini A, Sahoo S, Brown BD, Hajjar RJ, Kho C. miR-146a Suppresses SUMO1 Expression and Induces Cardiac Dysfunction in Maladaptive Hypertrophy. Circ Res 2019; 123:673-685. [PMID: 30355233 DOI: 10.1161/circresaha.118.312751] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RATIONALE Abnormal SUMOylation has emerged as a characteristic of heart failure (HF) pathology. Previously, we found reduced SUMO1 (small ubiquitin-like modifier 1) expression and SERCA2a (sarcoplasmic reticulum Ca2+-ATPase) SUMOylation in human and animal HF models. SUMO1 gene delivery or small molecule activation of SUMOylation restored SERCA2a SUMOylation and cardiac function in HF models. Despite the critical role of SUMO1 in HF, the regulatory mechanisms underlying SUMO1 expression are largely unknown. OBJECTIVE To examine miR-146a-mediated SUMO1 regulation and its consequent effects on cardiac morphology and function. METHODS AND RESULTS In this study, miR-146a was identified as a SUMO1-targeting microRNA in the heart. A strong correlation was observed between miR-146a and SUMO1 expression in failing mouse and human hearts. miR-146a was manipulated in cardiomyocytes through AAV9 (adeno-associated virus serotype 9)-mediated gene delivery, and cardiac morphology and function were analyzed by echocardiography and hemodynamics. Overexpression of miR-146a reduced SUMO1 expression, SERCA2a SUMOylation, and cardiac contractility in vitro and in vivo. The effects of miR-146a inhibition on HF pathophysiology were examined by transducing a tough decoy of miR-146a into mice subjected to transverse aortic constriction. miR-146a inhibition improved cardiac contractile function and normalized SUMO1 expression. The regulatory mechanisms of miR-146a upregulation were elucidated by examining the major miR-146a-producing cell types and transfer mechanisms. Notably, transdifferentiation of fibroblasts triggered miR-146a overexpression and secretion through extracellular vesicles, and the extracellular vesicle-associated miR-146a transfer was identified as the causative mechanism of miR-146a upregulation in failing cardiomyocytes. Finally, extracellular vesicles isolated from failing hearts were shown to contain high levels of miR-146a and exerted negative effects on the SUMO1/SERCA2a signaling axis and hence cardiomyocyte contractility. CONCLUSIONS Taken together, our results show that miR-146a is a novel regulator of the SUMOylation machinery in the heart, which can be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Jae Gyun Oh
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Shin Watanabe
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Ahyoung Lee
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Przemek A Gorski
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Philyoung Lee
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Dongtak Jeong
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Lifan Liang
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Yaxuan Liang
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Alessia Baccarini
- Department of Genetics and Genomic Sciences (A.B., B.D.B.), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York
| | - Susmita Sahoo
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Brian D Brown
- Department of Genetics and Genomic Sciences (A.B., B.D.B.), Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York
| | - Roger J Hajjar
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| | - Changwon Kho
- From the Department of Cardiology, Cardiovascular Research Center (J.G.O., S.W., A.L., P.A.G., P.L., D.J., L.L., Y.L., S.S., R.J.H., C.K.)
| |
Collapse
|
59
|
Cai J, Liu Z, Huang X, Shu S, Hu X, Zheng M, Tang C, Liu Y, Chen G, Sun L, Liu H, Liu F, Cheng J, Dong Z. The deacetylase sirtuin 6 protects against kidney fibrosis by epigenetically blocking β-catenin target gene expression. Kidney Int 2019; 97:106-118. [PMID: 31787254 DOI: 10.1016/j.kint.2019.08.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/06/2019] [Accepted: 08/22/2019] [Indexed: 01/20/2023]
Abstract
Fibrosis is a common pathologic pathway of progressive kidney disease involving complex signaling networks. The deacetylase sirtuin 6 (sirt6) was recently implicated in kidney injury. However, it remains elusive whether and how sirt6 contributes to the regulation of kidney fibrosis. Here, we demonstrate that sirt6 protects against kidney interstitial fibrosis through epigenetic regulation of β-catenin signaling. Sirt6 is markedly upregulated during fibrogenesis following obstructed nephropathy and kidney ischemia-reperfusion injury. Pharmacological inhibition of sirt6 deacetylase activity aggravates kidney fibrosis in obstructed nephropathy. Consistently, knockdown of sirt6 in mouse kidney proximal tubular epithelial cells aggravates transforming growth factor-β-induced fibrosis in vitro. Mechanistically, sirt6 deficiency results in augmented expression of the downstream target proteins of β-catenin signaling. We further show that sirt6 interacts with β-catenin during transforming growth factor-β treatment and binds to the promoters of β-catenin target genes, resulting in the deacetylation of histone H3K56 to prevent the transcription of fibrosis-related genes. Thus, our data reveal the anti-fibrotic function of sirt6 by epigenetically attenuating β-catenin target gene expression.
Collapse
Affiliation(s)
- Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China.
| | - Zhiwen Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Xian Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoqun Shu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Xiaoru Hu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Meiling Zheng
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Guochun Chen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, Hunan, China.
| |
Collapse
|
60
|
Chan HL, Morey L. Emerging Roles for Polycomb-Group Proteins in Stem Cells and Cancer. Trends Biochem Sci 2019; 44:688-700. [PMID: 31085088 DOI: 10.1016/j.tibs.2019.04.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 02/07/2023]
Abstract
Polycomb-group (PcG) complexes are multiprotein, evolutionarily conserved epigenetic machineries that regulate stem cell fate decisions and development, and are also implicated in cancer and other maladies. The PcG machinery can be divided into two major complexes: Polycomb repressive complex 1 and 2 (PRC1 and PRC2). Traditionally, PcG complexes have been associated with maintenance of gene repression mainly via histone-modifying activities. However, during the last years, increasing evidence indicates that the PcG complexes can also positively regulate gene transcription and modify non-histone substrates in multiple biological processes, cellular stages, and cancers. In this review, we will illustrate recent findings in PcG-mediated gene regulation, with special focus on the recently described non-classical functions of PcG complexes in stem cells and cancer.
Collapse
Affiliation(s)
- Ho Lam Chan
- Sylvester Comprehensive Cancer Center, Biomedical Research Building, 1501 NW 10th Avenue, Miami, FL 33136, USA; Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lluis Morey
- Sylvester Comprehensive Cancer Center, Biomedical Research Building, 1501 NW 10th Avenue, Miami, FL 33136, USA; Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
61
|
Protective role of the deSUMOylating enzyme SENP3 in myocardial ischemia-reperfusion injury. PLoS One 2019; 14:e0213331. [PMID: 30973885 PMCID: PMC6459529 DOI: 10.1371/journal.pone.0213331] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/02/2019] [Indexed: 12/26/2022] Open
Abstract
Interruption of blood supply to the heart is a leading cause of death and disability. However, the molecular events that occur during heart ischemia, and how these changes prime consequent cell death upon reperfusion, are poorly understood. Protein SUMOylation is a post-translational modification that has been strongly implicated in the protection of cells against a variety of stressors, including ischemia-reperfusion. In particular, the SUMO2/3-specific protease SENP3 has emerged as an important determinant of cell survival after ischemic infarct. Here, we used the Langendorff perfusion model to examine changes in the levels and localisation of SUMOylated target proteins and SENP3 in whole heart. We observed a 50% loss of SENP3 from the cytosolic fraction of hearts after preconditioning, a 90% loss after ischemia and an 80% loss after ischemia-reperfusion. To examine these effects further, we performed ischemia and ischemia-reperfusion experiments in the cardiomyocyte H9C2 cell line. Similar to whole hearts, ischemia induced a decrease in cytosolic SENP3. Furthermore, shRNA-mediated knockdown of SENP3 led to an increase in the rate of cell death upon reperfusion. Together, our results indicate that cardiac ischemia dramatically alter levels of SENP3 and suggest that this may a mechanism to promote cell survival after ischemia-reperfusion in heart.
Collapse
|
62
|
Chen XL, Wang SF, Liang XT, Liang HX, Wang TT, Wu SQ, Qiu ZJ, Zhan R, Xu ZS. SENP2 exerts an anti‑tumor effect on chronic lymphocytic leukemia cells through the inhibition of the Notch and NF‑κB signaling pathways. Int J Oncol 2018; 54:455-466. [PMID: 30431078 PMCID: PMC6317657 DOI: 10.3892/ijo.2018.4635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/19/2018] [Indexed: 01/28/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is one of the most often diagnosed hematological malignant tumors in the Western world and a type of inert B-cell lymphoma that commonly attacks the elderly. Small ubiquitin related modifier (SUMO)-specific protease 2 (SENP2) can act as a suppressor in various types of cancer by regulating the stability of β-catenin to affect the Notch signaling pathway; however, it has a low expression level in CLL cells. In this study, we firstly used western blot analysis and RT-qPCR to detect the protein and mRNA expression levels of SENP2 in the peripheral blood of patients with CLL and healthy volunteers. Secondly, we overexpressed or knocked down the expression of SENP2 in CLL cells and then determined the cell invasive and chemotactic ability in a Transwell assay and chemotaxis assay. We examined the sensitivity of the cells to cytarabine and dexamethasone via a CCK-8 assay and determined the cell apoptotic condition and the expression of the Notch signaling pathway using flow cytometry and western blot analysis. The results demonstrated that the patients with CLL had relatively low expression levels of SENP2. The overexpression of SENP2 in the CLL cells decreased their invasive and proliferative ability, as well as their chemotactic response and enhanced their sensitivity to cytarabine and dexamethasone, while it promoted cell apoptosis. The silencing of SENP2 in the CLL cells generally produced the opposite results. We thus hypothesized that the overexpression of SENP2 downregulated β-catenin expression, thus inhibiting the Notch signaling pathway in CLL cells. Moreover, the nuclear factor (NF)-κB signaling pathway was also regulated by the overexpression of SENP2. On the whole, the findings of this study indicate tha SENP2 can act as a tumor suppressor in CLL cells, and may thus prove to be a novel target for CLL treatment in clinical practice.
Collapse
Affiliation(s)
- Xiu-Li Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Shi-Fen Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xue-Ting Liang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Hui-Xin Liang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Ting-Ting Wang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Shun-Quan Wu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Zong-Jian Qiu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Rong Zhan
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Zhen-Shu Xu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
63
|
Peng Y, Zhang X, Lin H, Deng S, Huang Y, Qin Y, Feng X, Yan R, Zhao Y, Cheng Y, Wei Y, Wang J, Chen W, Fan X, Ashktorab H, Smoot D, Meltzer SJ, Li S, Zhang Z, Jin Z. Inhibition of miR‑194 suppresses the Wnt/β‑catenin signalling pathway in gastric cancer. Oncol Rep 2018; 40:3323-3334. [PMID: 30542715 PMCID: PMC6196585 DOI: 10.3892/or.2018.6773] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 10/01/2018] [Indexed: 01/01/2023] Open
Abstract
A mounting body of evidence has revealed that microRNAs (miRs) serve pivotal roles in various developmental processes, and in tumourigenesis, by binding to target genes and subsequently regulating gene expression. Continued activation of the Wnt/β-catenin signalling is positively associated with human malignancy. In addition, miR-194 dysregulation has been implicated in gastric cancer (GC); however, the molecular mechanisms underlying the effects of miR-194 on GC carcinogenesis remain to be elucidated. The present study demonstrated that miR-194 was upregulated in GC tissues and SUFU negative regulator of Ηedgehog signaling (SUFU) was downregulated in GC cell lines. Subsequently, inhibition of miR-194 attenuated nuclear accumulation of β-catenin, which consequently blocked Wnt/β-catenin signalling. In addition, the cytoplasmic translocation of β-catenin induced by miR-194 inhibition was mediated by SUFU. Furthermore, genes associated with the Wnt/β-catenin signalling pathway were revealed to be downregulated following inhibition of the Wnt signalling pathway by miR-194 suppression. Finally, the results indicated that cell apoptosis was markedly increased in response to miR-194 inhibition, strongly suggesting the carcinogenic effects of miR-194 in GC. Taken together, these findings demonstrated that miR-194 may promote gastric carcinogenesis through activation of the Wnt/β-catenin signalling pathway, making it a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Yin Peng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Xiaojing Zhang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Huijuan Lin
- Department of Pathology and Pathophysiology, The Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Shiqi Deng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Yong Huang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Xianling Feng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Ruibin Yan
- Laboratory of Chemical Genomics, The Shenzhen Graduate School of Peking University, Shenzhen, Guangdong 518055, P.R. China
| | - Yanqiu Zhao
- Laboratory of Chemical Genomics, The Shenzhen Graduate School of Peking University, Shenzhen, Guangdong 518055, P.R. China
| | - Yulan Cheng
- Department of Medicine/GI Division, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Yanjie Wei
- Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong 518000, P.R. China
| | - Jian Wang
- Department of Pathology and Pathophysiology, The Guangzhou Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Wangchun Chen
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Xinmin Fan
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| | - Hassan Ashktorab
- Department of Medicine and Cancer Center, Howard University, College of Medicine, Washington, DC 20060, USA
| | - Duane Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN 37208, USA
| | - Stephen J Meltzer
- Department of Medicine/GI Division, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Song Li
- Laboratory of Chemical Genomics, The Shenzhen Graduate School of Peking University, Shenzhen, Guangdong 518055, P.R. China
| | - Zhong Zhang
- Department of Pathology, College of Basic Medical Sciences, Shenyang Medical College, Shenyang, Liaoning 110034, P.R. China
| | - Zhe Jin
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pathology, The Shenzhen University School of Medicine, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
64
|
Rodríguez-Castañeda F, Lemma RB, Cuervo I, Bengtsen M, Moen LM, Ledsaak M, Eskeland R, Gabrielsen OS. The SUMO protease SENP1 and the chromatin remodeler CHD3 interact and jointly affect chromatin accessibility and gene expression. J Biol Chem 2018; 293:15439-15454. [PMID: 30082317 DOI: 10.1074/jbc.ra118.002844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/12/2018] [Indexed: 01/22/2023] Open
Abstract
The small ubiquitin-like modifier (SUMO) post-translationally modifies lysine residues of transcription factors and co-regulators and thereby contributes to an important layer of control of the activities of these transcriptional regulators. Likewise, deSUMOylation of these factors by the sentrin-specific proteases (SENPs) also plays a role in gene regulation, but whether SENPs functionally interact with other regulatory factors that control gene expression is unclear. In the present work, we focused on SENP1, specifically, on its role in activation of gene expression investigated through analysis of the SENP1 interactome, which revealed that SENP1 physically interacts with the chromatin remodeler chromodomain helicase DNA-binding protein 3 (CHD3). Using several additional methods, including GST pulldown and co-immunoprecipitation assays, we validated and mapped this interaction, and using CRISPR-Cas9-generated CHD3- and SENP1-KO cells (in the haploid HAP1 cell line), we investigated whether these two proteins are functionally linked in regulating chromatin remodeling and gene expression. Genome-wide ATAC-Seq analysis of the CHD3- and SENP1-KO cells revealed a large degree of overlap in differential chromatin openness between these two mutant cell lines. Moreover, motif analysis and comparison with ChIP-Seq profiles in K562 cells pointed to an association of CHD3 and SENP1 with CCCTC-binding factor (CTCF) and SUMOylated chromatin-associated factors. Lastly, genome-wide RNA-Seq also indicated that these two proteins co-regulate the expression of several genes. We propose that the functional link between chromatin remodeling by CHD3 and deSUMOylation by SENP1 uncovered here provides another level of control of gene expression.
Collapse
Affiliation(s)
| | - Roza Berhanu Lemma
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and
| | - Ignacio Cuervo
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and
| | - Mads Bengtsen
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and
| | - Lisa Marie Moen
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and
| | - Marit Ledsaak
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and
| | - Ragnhild Eskeland
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and.,the Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, P.O. Box 1112 Blindern, N-0317 Oslo, Norway
| | - Odd Stokke Gabrielsen
- From the Department of Biosciences, University of Oslo, P. O. Box 1066 Blindern, N-0316 Oslo and
| |
Collapse
|
65
|
Xiao N, Li H, Yu W, Gu C, Fang H, Peng Y, Mao H, Fang Y, Ni W, Yao M. SUMO‐specific protease 2 (SENP2) suppresses keratinocyte migration by targeting NDR1 for de‐SUMOylation. FASEB J 2018; 33:163-174. [DOI: 10.1096/fj.201800353r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ning Xiao
- Cancer Institute of Traditional Chinese MedicineLonghua HospitalShanghai University of Traditional Chinese Medicine Shanghai China
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and OncologyMinistry of Health and Pediatric Translational Medicine InstituteShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Weirong Yu
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Chuan Gu
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and OncologyMinistry of Health and Pediatric Translational Medicine InstituteShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Yinbo Peng
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Heshui Mao
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Yong Fang
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Wei Ni
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Min Yao
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
66
|
SUMO1/sentrin/SMT3 specific peptidase 2 modulates target molecules and its corresponding functions. Biochimie 2018; 152:6-13. [PMID: 29908207 DOI: 10.1016/j.biochi.2018.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 06/11/2018] [Indexed: 12/11/2022]
Abstract
Small ubiquitin-like modifier (SUMOylation) is a reversible post-translational modification, which plays important roles in numerous biological processes. SUMO could be covalently attached to target proteins in an isopeptide bond manner that occurs via a lysine ε-amino group on the target proteins and the glycine on SUMO C-terminus. This covalent binding could affect the subcellular localization and stability of target proteins. SUMO modification can be reversed by members of the Sentrin/SUMO-specific proteases (SENPs) family, which are highly evolutionarily conserved from yeast to human. SENP2, a member of the SENPs family, mainly plays a physiological function in the nucleus. SENP2 can promote maturity of the SUMO and deSUMOylate for single-SUMO modified or poly-SUMO modified proteins. SENP2 can affect the related biological processes through its peptidase activity or the amino terminal transcriptional repression domain. It plays important roles by inhibiting or activating some molecular functions. Therefore, the research achievements of SENP2 are reviewed in order to understand its related functions and the underlying molecular mechanisms and provide a clue for future research on SENP2.
Collapse
|
67
|
Domingues P, Hale BG. Functional Insights into ANP32A-Dependent Influenza A Virus Polymerase Host Restriction. Cell Rep 2018; 20:2538-2546. [PMID: 28903035 PMCID: PMC5608968 DOI: 10.1016/j.celrep.2017.08.061] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/02/2017] [Accepted: 08/18/2017] [Indexed: 01/25/2023] Open
Abstract
Host restriction of influenza A virus limits pandemic emergence. The viral RNA polymerase (vPol) is an essential enzyme that must adapt for avian viruses to replicate in humans. Species differences in host ANP32A dictate adaptation: human ANP32A lacks an uncharacterized 33 amino-acid insertion that is present in avian ANP32A. Here, we uncover important contributions of host SUMOylation to vPol activity, including avANP32A function. We also identify a hydrophobic SUMO interaction motif (SIM)-like sequence unique to avANP32A that critically supports avian-signature vPol. Unrelated SIM sequences partially recapitulate this function when introduced into huANP32A. By investigating ANP32A-vPol interactions, we find that huANP32A interacts weakly with both human- and avian-signature vPols, while the hydrophobic motif of avANP32A promotes stronger interactions. Furthermore, we identify a highly acidic stretch in avANP32A that constitutes a major site of vPol interaction. Our data suggest compensatory mechanisms underlying vPol adaptation to host ANP32A independent of species-specific interactions. Host SUMOylation contributes to influenza A virus polymerase (vPol) activity Avian ANP32A harbors a unique hydrophobic SUMO interaction motif-like sequence Avian ANP32A hydrophobic motif enhances vPol interaction to overcome restriction Interactions between ANP32A and vPol are independent of PB2-627 identity
Collapse
Affiliation(s)
- Patricia Domingues
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Benjamin G Hale
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
68
|
Maruyama T, Abe Y, Niikura T. SENP1 and SENP2 regulate SUMOylation of amyloid precursor protein. Heliyon 2018; 4:e00601. [PMID: 29862363 PMCID: PMC5968171 DOI: 10.1016/j.heliyon.2018.e00601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/19/2018] [Accepted: 04/05/2018] [Indexed: 11/29/2022] Open
Abstract
Amyloid β, a key molecule in the pathogenesis of Alzheimer's disease (AD), is produced from amyloid precursor protein (APP) by the cleavage of secretases. APP is SUMOylated near the cleavage site of β-secretase. SUMOylation of APP reduces amyloid β production, but its regulatory system is still unclear. SUMOylation, a modification at a lysine residue of a target protein, is mediated by activating, conjugating, and ligating enzymes and is reversed by a family of sentrin/SUMO-specific proteases (SENPs). Here, we found that both SENP1 and SENP2 induced de-SUMOylation of APP. Using quantitative PCR, we also found that expression of SENP1 but not SENP2 increased in an age-dependent manner only in female mice. The results of immunoblot analyses showed that the protein expression was consistent with the PCR results. Females, compared to males, have a higher incidence of AD in humans and show more aggressive amyloid pathology in AD mouse models. Our results provide a clue to understanding the role of SUMOylation in the sex difference in AD pathogenesis.
Collapse
Affiliation(s)
- Takuma Maruyama
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Japan
| | - Takako Niikura
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, Japan
| |
Collapse
|
69
|
Neddylation mediates ventricular chamber maturation through repression of Hippo signaling. Proc Natl Acad Sci U S A 2018; 115:E4101-E4110. [PMID: 29632206 DOI: 10.1073/pnas.1719309115] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
During development, ventricular chamber maturation is a crucial step in the formation of a functionally competent postnatal heart. Defects in this process can lead to left ventricular noncompaction cardiomyopathy and heart failure. However, molecular mechanisms underlying ventricular chamber development remain incompletely understood. Neddylation is a posttranslational modification that attaches ubiquitin-like protein NEDD8 to protein targets via NEDD8-specific E1-E2-E3 enzymes. Here, we report that neddylation is temporally regulated in the heart and plays a key role in cardiac development. Cardiomyocyte-specific knockout of NAE1, a subunit of the E1 neddylation activating enzyme, significantly decreased neddylated proteins in the heart. Mice lacking NAE1 developed myocardial hypoplasia, ventricular noncompaction, and heart failure at late gestation, which led to perinatal lethality. NAE1 deletion resulted in dysregulation of cell cycle-regulatory genes and blockade of cardiomyocyte proliferation in vivo and in vitro, which was accompanied by the accumulation of the Hippo kinases Mst1 and LATS1/2 and the inactivation of the YAP pathway. Furthermore, reactivation of YAP signaling in NAE1-inactivated cardiomyocytes restored cell proliferation, and YAP-deficient hearts displayed a noncompaction phenotype, supporting an important role of Hippo-YAP signaling in NAE1-depleted hearts. Mechanistically, we found that neddylation regulates Mst1 and LATS2 degradation and that Cullin 7, a NEDD8 substrate, acts as the ubiquitin ligase of Mst1 to enable YAP signaling and cardiomyocyte proliferation. Together, these findings demonstrate a role for neddylation in heart development and, more specifically, in the maturation of ventricular chambers and also identify the NEDD8 substrate Cullin 7 as a regulator of Hippo-YAP signaling.
Collapse
|
70
|
Yu F, Shi G, Cheng S, Chen J, Wu SY, Wang Z, Xia N, Zhai Y, Wang Z, Peng Y, Wang D, Du JX, Liao L, Duan SZ, Shi T, Cheng J, Chiang CM, Li J, Wong J. SUMO suppresses and MYC amplifies transcription globally by regulating CDK9 sumoylation. Cell Res 2018; 28:670-685. [PMID: 29588524 DOI: 10.1038/s41422-018-0023-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/28/2018] [Accepted: 02/11/2018] [Indexed: 01/21/2023] Open
Abstract
Regulation of transcription is fundamental to the control of cellular gene expression and function. Although recent studies have revealed a role for the oncoprotein MYC in amplifying global transcription, little is known as to how the global transcription is suppressed. Here we report that SUMO and MYC mediate opposite effects upon global transcription by controlling the level of CDK9 sumoylation. On one hand, SUMO suppresses global transcription via sumoylation of CDK9, the catalytic subunit of P-TEFb kinase essential for productive transcriptional elongation. On the other hand, MYC amplifies global transcription by antagonizing CDK9 sumoylation. Sumoylation of CDK9 blocks its interaction with Cyclin T1 and thus the formation of active P-TEFb complex. Transcription profiling analyses reveal that SUMO represses global transcription, particularly of moderately to highly expressed genes and by generating a sumoylation-resistant CDK9 mutant, we confirm that sumoylation of CDK9 inhibits global transcription. Together, our data reveal that SUMO and MYC oppositely control global gene expression by regulating the dynamic sumoylation and desumoylation of CDK9.
Collapse
Affiliation(s)
- Fang Yu
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guang Shi
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.,Key Laboratory of Gene Engineering of the Ministry of Education and Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shimeng Cheng
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiwei Chen
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shwu-Yuan Wu
- Simmons Comprehensive Cancer Center, Department of Biochemistry, and Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Zhiqiang Wang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Nansong Xia
- 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
| | - Yunhao Zhai
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhenxing Wang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yu Peng
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dong Wang
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - James X Du
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiology, Shanghai Research Institute of Stomatology, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tieliu Shi
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, 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
| | - Cheng-Ming Chiang
- Simmons Comprehensive Cancer Center, Department of Biochemistry, and Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Jiwen Li
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
71
|
Kunz K, Piller T, Müller S. SUMO-specific proteases and isopeptidases of the SENP family at a glance. J Cell Sci 2018; 131:131/6/jcs211904. [DOI: 10.1242/jcs.211904] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT
The ubiquitin-related SUMO system controls many cellular signaling networks. In mammalian cells, three SUMO forms (SUMO1, SUMO2 and SUMO3) act as covalent modifiers of up to thousands of cellular proteins. SUMO conjugation affects cell function mainly by regulating the plasticity of protein networks. Importantly, the modification is reversible and highly dynamic. Cysteine proteases of the sentrin-specific protease (SENP) family reverse SUMO conjugation in mammalian cells. In this Cell Science at a Glance article and the accompanying poster, we will summarize how the six members of the mammalian SENP family orchestrate multifaceted deconjugation events to coordinate cell processes, such as gene expression, the DNA damage response and inflammation.
Collapse
Affiliation(s)
- Kathrin Kunz
- Institute of Biochemistry II, Goethe University, Medical School, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Tanja Piller
- Institute of Biochemistry II, Goethe University, Medical School, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Stefan Müller
- Institute of Biochemistry II, Goethe University, Medical School, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
72
|
Bu FT, Chen Y, Yu HX, Chen X, Yang Y, Pan XY, Wang Q, Wu YT, Huang C, Meng XM, Li J. SENP2 alleviates CCl 4-induced liver fibrosis by promoting activated hepatic stellate cell apoptosis and reversion. Toxicol Lett 2018. [PMID: 29535048 DOI: 10.1016/j.toxlet.2018.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SUMOylation and deSUMOylation, a dynamic process, is proved to be involved in various fibrotic diseases. Here, we found SENP2, one of deSUMOylation protease family member, was decreased in CCl4-induced mice fibrotic liver tissues, primary HSCs and restored after spontaneously recovery. In addition, HSC-T6 cells with TGF-β1 treatment resulted in a significant reduction of SENP2. Ectopic expression of SENP2 hindered cells activation and proliferation induced by TGF-β1 while knockdown of SENP2 showed an opposite effect. Importantly, SENP2 promoted apoptosis of HSC-T6 cells activated by TGF-β1. Furthermore, restoration of SENP2 was observed in inactivated HSCs after adipogenic differentiation mixture (MDI) treatment. Inadequate SENP2 inhibited the reversion of HSC-T6 cells, featured as aberrant expressions of α-SMA and col1a1, two markers of liver fibrosis. It has been reported SENP2 was a suppressant regulator of Wnt/β-catenin signal pathway. Similarly, we found SENP2 has a negative effect on β-catenin as well as its downstream genes C-myc and CyclinD1 in liver fibrosis. Collectively, our data indicated SENP2 may be involved in HSCs apoptosis and reversion in liver fibrosis.
Collapse
Affiliation(s)
- Fang-Tian Bu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yu Chen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hai-Xia Yu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xin Chen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yang Yang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue-Yin Pan
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Qin Wang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yu-Ting Wu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Cheng Huang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| |
Collapse
|
73
|
Bernstock JD, Ye D, Smith JA, Lee YJ, Gessler FA, Yasgar A, Kouznetsova J, Jadhav A, Wang Z, Pluchino S, Zheng W, Simeonov A, Hallenbeck JM, Yang W. Quantitative high-throughput screening identifies cytoprotective molecules that enhance SUMO conjugation via the inhibition of SUMO-specific protease (SENP)2. FASEB J 2018; 32:1677-1691. [PMID: 29146736 PMCID: PMC5892725 DOI: 10.1096/fj.201700711r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/06/2017] [Indexed: 12/18/2022]
Abstract
The development of novel neuroprotective treatments for acute stroke has been fraught with failures, which supports the view of ischemic brain damage as a highly complex multifactorial process. Post-translational modifications such as small ubiquitin-like modifier (SUMO)ylation have emerged as critical molecular regulatory mechanisms in states of both homeostasis and ischemic stress, as evidenced by our previous work. Accordingly, the clinical significance of the selective control of the global SUMOylation process has become apparent in studies of ischemic pathobiology and pathophysiology. Herein, we describe a process capable of identifying and characterizing small molecules with the potential of targeting the SUMO system through inhibition of SUMO deconjugation in an effort to develop novel stroke therapies.-Bernstock, J. D., Ye, D., Smith, J. A., Lee, Y.-J., Gessler, F. A., Yasgar, A., Kouznetsova, J., Jadhav, A., Wang, Z., Pluchino, S., Zheng, W., Simeonov, A., Hallenbeck, J. M., Yang, W. Quantitative high-throughput screening identifies cytoprotective molecules that enhance SUMO-conjugation via the inhibition of SUMO-specific protease (SENP)2.
Collapse
Affiliation(s)
- Joshua D. Bernstock
- Stroke Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, Maryland, USA
- Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute and National Institute of Health Research Biomedical Research Centre, University of Cambridge, United Kingdom
- UAB School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Daniel Ye
- Stroke Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, Maryland, USA
| | - Jayden A. Smith
- Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute and National Institute of Health Research Biomedical Research Centre, University of Cambridge, United Kingdom
| | - Yang-Ja Lee
- Stroke Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, Maryland, USA
| | - Florian A. Gessler
- Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute and National Institute of Health Research Biomedical Research Centre, University of Cambridge, United Kingdom
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Jennifer Kouznetsova
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Zhuoran Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Stefano Pluchino
- Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute and National Institute of Health Research Biomedical Research Centre, University of Cambridge, United Kingdom
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA; and
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA; and
| | - John M. Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, Maryland, USA
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
74
|
Chen X, Zhang S, Huang J, Dong W, Xiao H, Shao H, Cheng J, Wu H, Qi Y. Hyper-SUMOylation of K + Channels in Sudden Unexplained Death in Epilepsy: Isolation and Primary Culture of Dissociated Hippocampal Neurons from Newborn Mice for Subcellular Localization. Methods Mol Biol 2018; 1684:63-71. [PMID: 29058184 DOI: 10.1007/978-1-4939-7362-0_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The physiological characteristics of rat and murine hippocampal neurons are widely studied, especially because of the involvement of the hippocampus in learning, memory, and neurological functions. Primary cultures of hippocampal neurons are commonly used to discover cellular and molecular mechanisms in neurobiology. By isolating and culturing individual hippocampal neurons, neuroscientists are able to investigate the activity of neurons at the individual cell and single synapse level, and to analyze properties related to cellular structure, cellular trafficking, and individual protein subcellular localization or protein-protein interaction using a variety of biochemical techniques. Conclusions addressed from such research are critical for testing theories related to memory, learning, and neurological functions. Here, we will describe how to isolate and culture primary hippocampal cells from newborn mice. The hippocampus may be isolated from newborn mice in as short as 2 min, and the cell cultures can be maintained for up to 2 weeks, and then ready for investigation of subcellular localization of K+ channel proteins and interaction with SUMO-specific protease 2 (SENP2). The protocol provides a fast and efficient technique for the culture of neuronal cells from mouce hippocampal tissue, and will ensure the immunocytochemistry detection of subcellular localization or protein-protein interactions in neurological research.
Collapse
Affiliation(s)
- Xu Chen
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Shanshan Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Jifang Huang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Wanying Dong
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Hui Xiao
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Huanjie Shao
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongmei Wu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| | - Yitao Qi
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| |
Collapse
|
75
|
Lin FM, Kumar S, Ren J, Karami S, Bahnassy S, Li Y, Zheng X, Wang J, Bawa-Khalfe T. SUMOylation of HP1α supports association with ncRNA to define responsiveness of breast cancer cells to chemotherapy. Oncotarget 2017; 7:30336-49. [PMID: 27107417 PMCID: PMC5058684 DOI: 10.18632/oncotarget.8733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/03/2016] [Indexed: 01/19/2023] Open
Abstract
Epigenetic reprogramming allows cancer cells to bypass normal checkpoints and potentiate aberrant proliferation. Several chromatin regulators are subject to reversible SUMO-modification but little is known about how SUMOylation of chromatin-remodelers modulates the cancer epigenome. Recently, we demonstrated that SUMO-protease SENP7L is upregulated in aggressive BCa and maintains hypoSUMOylated heterochromatin protein 1-α (HP1α). Canonical models define HP1α as a "reader" of repressive H3K9m3 marks that supports constitutive heterochromatin. It is unclear how SUMOylation affects HP1α function in BCa cells. This report shows HP1α SUMO-dynamics are closely regulated in a complex with SENP7L and SUMO-E3 Polycomb-2 (PC2/CBX4). This complex accumulates at H3K9m3 sites, hypoSUMOylates HP1α and PC2, and reduces PC2's SUMO-E3 activity. HyperSUMO conditions cause complex dissociation, SUMOylation of PC2 and HP1α, and recruitment of SUMOylated HP1α to multiple DNA-repair genes including Rad51C. SUMOylated HP1α's enrichment at euchromatin requires chromatin-bound non-coding RNA (ncRNA), reduces Rad51C protein, and increases DNA-breaks in BCa cells. Hence, HP1α SUMOylation and consistently low SENP7L increase efficacy of DNA-damaging chemotherapeutic agents. BCa patients on chemotherapy that express low SENP7L exhibit greater survival rates than patients with high SENP7L. Collectively, these studies suggest that SUMOylated HP1α is a critical epigenetic-regulator of DNA-repair in BCa that could define chemotherapy responsiveness.
Collapse
Affiliation(s)
- Feng-Ming Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Santosh Kumar
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Jing Ren
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Samaneh Karami
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Shaymaa Bahnassy
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Yue Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tasneem Bawa-Khalfe
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
76
|
Aberrant GATA2 epigenetic dysregulation induces a GATA2/GATA6 switch in human gastric cancer. Oncogene 2017; 37:993-1004. [PMID: 29106391 DOI: 10.1038/onc.2017.397] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/08/2017] [Accepted: 09/15/2017] [Indexed: 02/07/2023]
Abstract
Six GATA transcription factors play important roles in eukaryotic development. Among these, GATA2, an essential factor for the hematopoietic cell lineage, exhibits low expression in human gastric tissues, whereas GATA6, which is crucial for gastrointestinal development and differentiation, is frequently amplified and/or overexpressed in human gastric cancer. Interestingly, we found that GATA6 was overexpressed in human gastric cancer cells only when GATA2 expression was completely absent, thereby showing an inverse correlation between GATA2 and GATA6. In gastric cancer cells that express high GATA6 levels, a GATA2 CpG island is hypermethylated, repressing expression in these cells. In contrast, GATA6 expression is undetectable in GATA2-overexpressing gastric cancer cells, which lack GATA2 DNA methylation. Furthermore, PRC2 complex-mediated transcriptional silencing of GATA6 was observed in the GATA2-overexpressing cells. We also show that the GATA2 and PRC2 complexes are enriched within the GATA6 locus, and that the recruitment of the PRC2 complex is impaired by disrupting GATA2 expression, resulting in GATA6 upregulation. In addition, ectopic GATA2 expression significantly downregulates GATA6 expression, suggesting GATA2 directly represses GATA6. Furthermore, GATA6 downregulation showed antitumor activity by inducing growth arrest. Finally, we show that aberrant GATA2 methylation occurs early during the multistep process of gastric carcinogenesis regardless of Helicobacter pylori infection. Taken together, GATA2 dysregulation by epigenetic modification is associated with unfavorable phenotypes in human gastric cancer cells by allowing GATA6 expression.
Collapse
|
77
|
Knittle AM, Helkkula M, Johnson MS, Sundvall M, Elenius K. SUMOylation regulates nuclear accumulation and signaling activity of the soluble intracellular domain of the ErbB4 receptor tyrosine kinase. J Biol Chem 2017; 292:19890-19904. [PMID: 28974580 DOI: 10.1074/jbc.m117.794271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/22/2017] [Indexed: 01/24/2023] Open
Abstract
Erb-B2 receptor tyrosine kinase 4 (ErbB4) is a kinase that can signal via a proteolytically released intracellular domain (ICD) in addition to classical receptor tyrosine kinase-activated signaling cascades. Previously, we have demonstrated that ErbB4 ICD is posttranslationally modified by the small ubiquitin-like modifier (SUMO) and functionally interacts with the PIAS3 SUMO E3 ligase. However, direct evidence of SUMO modification in ErbB4 signaling has remained elusive. Here, we report that the conserved lysine residue 714 in the ErbB4 ICD undergoes SUMO modification, which was reversed by sentrin-specific proteases (SENPs) 1, 2, and 5. Although ErbB4 kinase activity was not necessary for the SUMOylation, the SUMOylated ErbB4 ICD was tyrosine phosphorylated to a higher extent than unmodified ErbB4 ICD. Mutation of the SUMOylation site compromised neither ErbB4-induced phosphorylation of the canonical signaling pathway effectors Erk1/2, Akt, or STAT5 nor ErbB4 stability. In contrast, SUMOylation was required for nuclear accumulation of the ErbB4 ICD. We also found that Lys-714 was located within a leucine-rich stretch, which resembles a nuclear export signal, and could be inactivated by site-directed mutagenesis. Furthermore, SUMOylation modulated the interaction of ErbB4 with chromosomal region maintenance 1 (CRM1), the major nuclear export receptor for proteins. Finally, the SUMO acceptor lysine was functionally required for ErbB4 ICD-mediated inhibition of mammary epithelial cell differentiation in a three-dimensional cell culture model. Our findings indicate that a SUMOylation-mediated mechanism regulates nuclear localization and function of the ICD of ErbB4 receptor tyrosine kinase.
Collapse
Affiliation(s)
- Anna M Knittle
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland.,Turku Doctoral Programmes of Biomedical Sciences and Molecular Medicine, University of Turku, FI-20014 Turku, Finland
| | - Maria Helkkula
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, FI-20500 Turku, Finland, and
| | - Maria Sundvall
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland, .,Department of Oncology and Radiotherapy, University of Turku and Turku University Hospital, FI-20014 Turku, Finland
| | - Klaus Elenius
- From the Department of Medical Biochemistry and Genetics, and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland, .,Department of Oncology and Radiotherapy, University of Turku and Turku University Hospital, FI-20014 Turku, Finland
| |
Collapse
|
78
|
Abstract
Many of the known SUMO substrates are nuclear proteins, which regulate gene expression and chromatin dynamics. Sumoylation, in general, appears to correlate with decreased transcriptional activity, and in many cases modulation of the chromatin template is implicated. Sumoylation of the core histones is associated with transcriptional silencing, and transcription factor sumoylation can decrease gene expression by promoting recruitment of chromatin modifying enzymes. Additionally, sumoylation of transcriptional corepressors and chromatin remodeling enzymes can influence interactions with other transcriptional regulators, and alter their enzymatic activity. In some cases, proteins that are components of transcriptional corepressor complexes have been shown to be SUMO E3 ligases, further emphasizing the integration of sumoylation with the regulation of chromatin remodeling. Despite the evidence suggesting that sumoylation is primarily repressive for access to chromatin, recent analyses suggest that protein sumoylation on the chromatin template may play important roles at highly expressed genes. Elucidating the dynamic interplay of sumoylation with other post-translational modifications of histones and chromatin associated proteins will be key to fully understanding the regulation of access to the chromatin template.
Collapse
|
79
|
Carranco R, Prieto-Dapena P, Almoguera C, Jordano J. SUMO-Dependent Synergism Involving Heat Shock Transcription Factors with Functions Linked to Seed Longevity and Desiccation Tolerance. FRONTIERS IN PLANT SCIENCE 2017; 8:974. [PMID: 28659940 PMCID: PMC5468958 DOI: 10.3389/fpls.2017.00974] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/23/2017] [Indexed: 05/03/2023]
Abstract
A transcriptional synergism between HaHSFA9 (A9) and HaHSFA4a (A4a) contributes to determining longevity and desiccation tolerance of sunflower (Helianthus annuus, L.) seeds. Potential lysine SUMOylation sites were identified in A9 and A4a and mutated to arginine. We show that A9 is SUMOylated in planta at K38. Although we did not directly detect SUMOylated A4a in planta, we provide indirect evidence from transient expression experiments indicating that A4a is SUMOylated at K172. Different combinations of wild type and SUMOylation site mutants of A9 and A4a were analyzed by transient expression in sunflower embryos and leaves. Although most of the precedents in literature link SUMOylation with repression, the A9 and A4a synergism was fully abolished when the mutant forms for both factors were combined. However, the combination of mutant forms of A9 and A4a did not affect the nuclear retention of A4a by A9; therefore, the analyzed mutations would affect the synergism after the mutual interaction and nuclear co-localization of A9 and A4a. Our results suggest a role for HSF SUMOylation during late, zygotic, embryogenesis. The SUMOylation of A9 (or A4a) would allow a crucial, synergic, transcriptional effect that occurs in maturing sunflower seeds.
Collapse
Affiliation(s)
| | | | | | - Juan Jordano
- Departamento de Biotecnología Vegetal, Instituto de Recursos Naturales y Agrobiología de Sevilla, Consejo Superior de Investigaciones CientíficasSeville, Spain
| |
Collapse
|
80
|
Wen B, Yuan H, Liu X, Wang H, Chen S, Chen Z, de The H, Zhou J, Zhu J. GATA5 SUMOylation is indispensable for zebrafish cardiac development. Biochim Biophys Acta Gen Subj 2017; 1861:1691-1701. [PMID: 28285006 DOI: 10.1016/j.bbagen.2017.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/07/2017] [Accepted: 03/07/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND SUMOylation is a critical regulatory protein modification in eukaryotic cells and plays a pivotal role in cardiac development and disease. Several cardiac transcription factors are modified by SUMO, but little is known about the impact of SUMOylation on their function during cardiac development. METHODS We used a zebrafish model to address the impact of SUMOylation on GATA5, an essential transcription factor in zebrafish cardiac development. GATA5 SUMOylation was probed by western blot, the subcellular localization and transcriptional activity of GATA5 mutants were examined by immunostaining and luciferase reporter assay. The in vivo function of GATA5 SUMOylation was evaluated by gata5 mutants mRNA microinjection and in situ hybridization in gata5 morphants and ubc9 mutants. RESULTS Firstly, we identified GATA5 as a SUMO substrate, and lysine 324 (K324) and lysine 360 (K360) as two major modification sites. Conversion of lysine to arginine at these two sites did not affect subcellular localization, but did affect the transcriptional activity of GATA5. Secondly, in vivo experiments demonstrated that the wild type (WT) and K324R mutant of gata5 could rescue impaired cardiac precursor differentiation, while the K360R mutant of gata5 drastically lost this potency in gata5 morphant. Furthermore, in SUMOylation-deficient ubc9 mutants, the abnormal expression pattern displayed by the early markers of cardiac development (nkx2.5 and mef2cb) could be restored using a sumo-gata5 fusion, but not with a WT gata5. CONCLUSION GATA5 SUMOylation is indispensable for early zebrafish cardiac development. GENERAL SIGNIFICANCE Our studies highlight the potential importance of transcription factor SUMOylation in cardiac development.
Collapse
Affiliation(s)
- Bin Wen
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yuan
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Liu
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haihong Wang
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Saijuan Chen
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhu Chen
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hugues de The
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée No. 11 Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris, France
| | - Jun Zhou
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Zhu
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée No. 11 Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris, France.
| |
Collapse
|
81
|
Monribot-Villanueva J, Zurita M, Vázquez M. Developmental transcriptional regulation by SUMOylation, an evolving field. Genesis 2017; 55. [PMID: 27935206 DOI: 10.1002/dvg.23009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 02/05/2023]
Abstract
SUMOylation is a reversible post-translational protein modification that affects the intracellular localization, stability, activity, and interactions of its protein targets. The SUMOylation pathway influences several nuclear and cytoplasmic processes. The expression of many genes, in particular those involved in development is finely tuned in space and time by several groups of proteins. There is growing evidence that transcriptional regulation mechanisms involve direct SUMOylation of transcriptional-related proteins such as initiation and elongation factors, and subunits of chromatin modifier and remodeling complexes originally described as members of the trithorax and Polycomb groups in Drosophila. Therefore, it is being unveiled that SUMOylation has a role in both, gene silencing and gene activation mechanisms. The goal of this review is to discuss the information on how SUMO modification in components of these multi-subunit complexes may have an effect in genome architecture and function and, therefore, in the regulation of gene expression in time and space.
Collapse
Affiliation(s)
- Juan Monribot-Villanueva
- Departamento de Fisiología Molecular y Genética del Desarrollo, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Mario Zurita
- Departamento de Fisiología Molecular y Genética del Desarrollo, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Martha Vázquez
- Departamento de Fisiología Molecular y Genética del Desarrollo, Instituto de Biotecnología-Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
82
|
Sumoylation in Development and Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:197-214. [DOI: 10.1007/978-3-319-50044-7_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
83
|
Pauws E, Stanier P. Sumoylation in Craniofacial Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:323-335. [PMID: 28197921 DOI: 10.1007/978-3-319-50044-7_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Craniofacial development requires a complex series of coordinated and finely tuned events to take place, during a relatively short time frame. These events are set in motion by switching on and off transcriptional cascades that involve the use of numerous signalling pathways and a multitude of factors that act at the site of gene transcription. It is now well known that amidst the subtlety of this process lies the intricate world of protein modification, and the posttranslational addition of the small ubiquitin -like modifier, SUMO, is an example that has been implicated in this process. Many proteins that are required for formation of various structures in the embryonic head and face adapt specific functions with SUMO modification. Interestingly, the main clinical phenotype reported for a disruption of the SUMO1 locus is the common birth defect cleft lip and palate. In this chapter therefore, we discuss the role of SUMO1 in craniofacial development, with emphasis on orofacial clefts. We suggest that these defects can be a sensitive indication of down regulated SUMO modification at a critical stage during embryogenesis. As well as specific mutations affecting the ability of particular proteins to be sumoylated, non-genetic events may have the effect of down-regulating the SUMO pathway to give the same result. Enzymes regulating the SUMO pathway may become important therapeutic targets in the preventative and treatment therapies for craniofacial defects in the future.
Collapse
Affiliation(s)
- Erwin Pauws
- Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Philip Stanier
- Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
84
|
Liu L, Yan X, Kong X, Zhao Y, Gong Z, Jin JB, Guo Y. Transcriptional Gene Silencing Maintained by OTS1 SUMO Protease Requires a DNA-Dependent Polymerase V-Dependent Pathway. PLANT PHYSIOLOGY 2017; 173:655-667. [PMID: 27852949 PMCID: PMC5210737 DOI: 10.1104/pp.16.01365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/14/2016] [Indexed: 05/21/2023]
Abstract
The expression of genes with aberrant structure is prevented at both the transcriptional and posttranscriptional regulation levels. Aberrant gene silencing at the posttranscriptional level is well studied; however, it is not well understood how aberrant genes are silenced at the transcriptional level. In this study, through genetic screening a transgenic report line that harbors an aberrant gene (35S-LUC, lacking 3'-untranslated region [3'-UTR]) and lacks luciferase (LUC) activity, we identify that the small ubiquitin-like modifier (SUMO) protease OTS1 gene is required for maintaining the silence of the reporter 35S-LUC and an endogenous mutator-like element MULE-F19G14 at the transcriptional level, which requires DNA-dependent RNA polymerase (Pol) V and DDR complex, but not Pol IV. The increased transcripts in ots1 mutants are terminated by the 3'-UTRs of downstream genes. In addition to ots1 mutations, mutations in several known or putative SUMO proteases and two SUMO E3 ligases, SIZ1 and MMS21, have similar effects on this silencing regulation. Taken together, our results reveal that the enzymes involved in the SUMOylation process restrain aberrant gene transcription by using a downstream gene 3'-UTR, and this regulation requires a functional Pol V-dependent pathway in Arabidopsis (Arabidopsis thaliana).
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| | - Xiaojing Yan
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| | - Xiangxiong Kong
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| | - Yiqiang Zhao
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| | - Zhizhong Gong
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| | - Jing Bo Jin
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| | - Yan Guo
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China (L.L., X.Y., Y.Z., Z.G., Y.G.); and
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China (X.K., J.B.J.)
| |
Collapse
|
85
|
Wang X, Li L, Wu Y, Zhang R, Zhang M, Liao D, Wang G, Qin G, Xu RH, Kang T. CBX4 Suppresses Metastasis via Recruitment of HDAC3 to the Runx2 Promoter in Colorectal Carcinoma. Cancer Res 2016; 76:7277-7289. [PMID: 27864346 DOI: 10.1158/0008-5472.can-16-2100] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/14/2016] [Accepted: 10/31/2016] [Indexed: 11/16/2022]
Abstract
Polycomb chromobox (CBX) proteins participate in the polycomb repressive complex (PRC1) that mediates epigenetic gene silencing and endows PRC1 with distinct oncogenic or tumor suppressor functions in a cell-type-dependent manner. In this study, we report that inhibition of cell migration, invasion, and metastasis in colorectal carcinoma requires CBX4-mediated repression of Runx2, a key transcription factor that promotes colorectal carcinoma metastasis. CBX4 inversely correlated with Runx2 expression in colorectal carcinoma tissues, and the combination of high CBX4 expression and low Runx2 expression significantly correlated with overall survival, more so than either CBX4 or Runx2 expression alone. Mechanistically, CBX4 maintained recruited histone deacetylase 3 (HDAC3) to the Runx2 promoter, which maintained a deacetylated histone H3K27 state to suppress Runx2 expression. This function of CBX4 was dependent on its interaction with HDAC3, but not on its SUMO E3 ligase, its chromodomain, or the PRC1 complex. Disrupting the CBX4-HDAC3 interaction abolished Runx2 inhibition as well as the inhibition of cell migration and invasion. Collectively, our data show that CBX4 may act as a tumor suppressor in colorectal carcinoma, and strategies that stabilize the interaction of CBX4 with HDAC3 may benefit the colorectal carcinoma patients with metastases. Cancer Res; 76(24); 7277-89. ©2016 AACR.
Collapse
Affiliation(s)
- Xin Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Liping Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Meifang Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dan Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ge Qin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
86
|
Yang W, Sheng H, Wang H. Targeting the SUMO pathway for neuroprotection in brain ischaemia. Stroke Vasc Neurol 2016; 1:101-107. [PMID: 28959470 PMCID: PMC5435206 DOI: 10.1136/svn-2016-000031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 08/05/2016] [Accepted: 08/06/2016] [Indexed: 12/20/2022] Open
Abstract
Small ubiquitin-like modifier (SUMO) conjugation (SUMOylation) is a post-translational protein modification that modulates almost all major cellular processes, and has been implicated in many human diseases. A growing body of evidence from in vitro and in vivo studies demonstrates that increasing global levels of SUMO conjugated proteins (global SUMOylation) protects cells against ischaemia-induced damage, while suppressing global SUMOylation promotes cell injury after ischaemia. Indeed, SUMOylation has emerged as a potential therapeutic target for neuroprotection in brain ischaemia, including global brain ischaemia and focal brain ischaemia (ischaemic stroke). Here, we summarise findings on the role of SUMOylation in human diseases, brain ischaemia in particular, and review recent developments in drug discovery targeting SUMOylation with a major focus on its neuroprotective applications.
Collapse
Affiliation(s)
- Wei Yang
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Huaxin Sheng
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Haichen Wang
- Multidisciplinary Neuroprotection Laboratories, Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
87
|
Da Silva-Ferrada E, Ribeiro-Rodrigues TM, Rodríguez MS, Girão H. Proteostasis and SUMO in the heart. Int J Biochem Cell Biol 2016; 79:443-450. [PMID: 27662810 DOI: 10.1016/j.biocel.2016.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022]
Abstract
Heart proteostasis relies on a complex and integrated network of molecular processes surveilling organ performance under physiological and pathological conditions. For this purpose, cardiac cells depend on the correct function of their proteolytic systems, such as the ubiquitin-proteasome system (UPS), autophagy and the calpain system. Recently, the role of protein SUMOylation (an ubiquitin-like modification), has emerged as important modulator of cardiac proteostasis, which will be the focus of this review.
Collapse
Affiliation(s)
- Elisa Da Silva-Ferrada
- Institute for Biomedical Imaging and Life Sciences (IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology (CNC), Institute for Biomedical Imaging and Life Sciences (IBILI) (CNC.IBILI), University of Coimbra, Coimbra, Portugal
| | - Teresa M Ribeiro-Rodrigues
- Institute for Biomedical Imaging and Life Sciences (IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology (CNC), Institute for Biomedical Imaging and Life Sciences (IBILI) (CNC.IBILI), University of Coimbra, Coimbra, Portugal
| | - Manuel S Rodríguez
- Institut des Technologies Avancées en Sciences du Vivant (ITAV), Université de Toulouse, CNRS, UPS, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, France
| | - Henrique Girão
- Institute for Biomedical Imaging and Life Sciences (IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology (CNC), Institute for Biomedical Imaging and Life Sciences (IBILI) (CNC.IBILI), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
88
|
Abstract
SUMOylation is a ubiquitin-related transient posttranslational modification pathway catalyzing the conjugation of small ubiquitin-like modifier (SUMO) proteins (SUMO1, SUMO2, and SUMO3) to lysine residues of proteins. SUMOylation targets a wide variety of cellular regulators and thereby affects a multitude of different cellular processes. SUMO/sentrin-specific proteases are able to remove SUMOs from targets, contributing to a tight control of SUMOylated proteins. Genetic and cell biological experiments indicate a critical role of balanced SUMOylation/deSUMOylation for proper cardiac development, metabolism, and stress adaptation. Here, we review the current knowledge about SUMOylation/deSUMOylation in the heart and provide an integrated picture of cardiac functions of the SUMO system under physiologic or pathologic conditions. We also describe potential therapeutic approaches targeting the SUMO machinery to combat heart disease.
Collapse
Affiliation(s)
- Luca Mendler
- From the Institute of Biochemistry II, Goethe University, Medical School, Frankfurt, Germany (L.M., S.M.); Institute of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary (L.M.); and Department I - Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.)
| | - Thomas Braun
- From the Institute of Biochemistry II, Goethe University, Medical School, Frankfurt, Germany (L.M., S.M.); Institute of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary (L.M.); and Department I - Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.).
| | - Stefan Müller
- From the Institute of Biochemistry II, Goethe University, Medical School, Frankfurt, Germany (L.M., S.M.); Institute of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary (L.M.); and Department I - Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (T.B.).
| |
Collapse
|
89
|
Lee A, Oh JG, Gorski PA, Hajjar RJ, Kho C. Post-translational Modifications in Heart Failure: Small Changes, Big Impact. Heart Lung Circ 2016; 25:319-24. [PMID: 26795636 PMCID: PMC4775300 DOI: 10.1016/j.hlc.2015.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/16/2015] [Accepted: 11/23/2015] [Indexed: 12/31/2022]
Abstract
Heart failure is a complex disease process with various aetiologies and is a significant cause of morbidity and death world-wide. Post-translational modifications (PTMs) alter protein structure and provide functional diversity in terms of physiological functions of the heart. In addition, alterations in protein PTMs have been implicated in human disease pathogenesis. Small ubiquitin-like modifier mediated modification (SUMOylation) pathway was found to play essential roles in cardiac development and function. Abnormal SUMOylation has emerged as a new feature of heart failure pathology. In this review, we will highlight the importance of SUMOylation as a regulatory mechanism of SERCA2a function, and its therapeutic potential for the treatment of heart failure.
Collapse
Affiliation(s)
- Ahyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jae Gyun Oh
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Przemek A Gorski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Changwon Kho
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
90
|
Huang C, Cheng J, Bawa-Khalfe T, Yao X, Chin YE, Yeh ETH. SUMOylated ORC2 Recruits a Histone Demethylase to Regulate Centromeric Histone Modification and Genomic Stability. Cell Rep 2016; 15:147-157. [PMID: 27052177 DOI: 10.1016/j.celrep.2016.02.091] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/17/2015] [Accepted: 02/26/2016] [Indexed: 01/25/2023] Open
Abstract
Origin recognition complex 2 (ORC2), a subunit of the ORC, is essential for DNA replication initiation in eukaryotic cells. In addition to a role in DNA replication initiation at the G1/S phase, ORC2 has been shown to localize to the centromere during the G2/M phase. Here, we show that ORC2 is modified by small ubiquitin-like modifier 2 (SUMO2), but not SUMO1, at the G2/M phase of the cell cycle. SUMO2-modification of ORC2 is important for the recruitment of KDM5A in order to convert H3K4me3 to H3K4me2, a "permissive" histone marker for α-satellite transcription at the centromere. Persistent expression of SUMO-less ORC2 led to reduced α-satellite transcription and impaired pericentric heterochromatin silencing, which resulted in re-replication of heterochromatin DNA. DNA re-replication eventually activated the DNA damage response, causing the bypass of mitosis and the formation of polyploid cells. Thus, ORC2 sustains genomic stability by recruiting KDM5A to maintain centromere histone methylation in order to prevent DNA re-replication.
Collapse
Affiliation(s)
- Chao Huang
- Texas Heart Institute, Houston, TX 77030, USA; Department of Cardiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; The Central Lab at Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinke Cheng
- Texas Heart Institute, Houston, TX 77030, USA; Department of Biochemistry and Molecular and Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tasneem Bawa-Khalfe
- Department of Cardiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Center for Nuclear Receptors and Cell Signaling and Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Xuebiao Yao
- Anhui Key Laboratory of Cellular Dynamics and Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei 230026, China
| | - Y Eugene Chin
- The Central Lab at Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Edward T H Yeh
- Texas Heart Institute, Houston, TX 77030, USA; Department of Cardiology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
91
|
Abstract
The GATA family of transcription factors consists of six proteins (GATA1-6) which are
involved in a variety of physiological and pathological processes. GATA1/2/3 are required
for differentiation of mesoderm and ectoderm-derived tissues, including the haematopoietic
and central nervous system. GATA4/5/6 are implicated in development and differentiation of
endoderm- and mesoderm-derived tissues such as induction of differentiation of embryonic
stem cells, cardiovascular embryogenesis and guidance of epithelial cell differentiation
in the adult.
Collapse
|
92
|
Cai J, Zuo Y, Wang T, Cao Y, Cai R, Chen FL, Cheng J, Mu J. A crucial role of SUMOylation in modulating Sirt6 deacetylation of H3 at lysine 56 and its tumor suppressive activity. Oncogene 2016; 35:4949-56. [PMID: 26898756 DOI: 10.1038/onc.2016.24] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/18/2015] [Accepted: 11/06/2015] [Indexed: 02/07/2023]
Abstract
Sirt6 is a histone deacetylase with NAD(+)-dependent activity. Sirt6 has been shown as a tumor suppressor partially via inhibiting the expression of c-Myc target genes and ribosome biogenesis. However, how to regulate Sirt6 activity is largely unknown. In this study, we identify that Sirt6 can be modified by small ubiquitin-like modifier. Sirt6 SUMOylation deficiency specifically decreases its deacetylation of H3K56 but not H3K9 in vivo. Mechanistically, we find that SUMOylation deficiency decreases Sirt6 binding with c-Myc, decreasing Sirt6 occupancy on the locus of c-Myc target genes. Therefore, Sirt6 SUMOylation deficiency reduces its deacetylation of H3k56 and its repression of c-Myc target genes. Moreover, Sirt6 SUMOylation deficiency reduces its suppression of cell proliferation and tumorigenesis. Thus, these results reveal that SUMOylation has an important role in regulation of Sirt6 deacetylation on H3K56, as well as its tumor suppressive activity.
Collapse
Affiliation(s)
- J Cai
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - T Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Cao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - R Cai
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - F-L Chen
- Shanghai Third People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - J Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - J Mu
- Department of Thoracic Surgery, Cancer Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| |
Collapse
|
93
|
Extraembryonic but not embryonic SUMO-specific protease 2 is required for heart development. Sci Rep 2016; 6:20999. [PMID: 26883797 PMCID: PMC4756675 DOI: 10.1038/srep20999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/15/2016] [Indexed: 02/01/2023] Open
Abstract
SUMO-specific protease 2 (SENP2) activities to remove SUMO from its substrates is essential for development of trophoblast stem cells, niches and lineages. Global deletion of SENP2 leads to midgestation lethality, and causes severe defects in the placenta which is accompanied by embryonic brain and heart abnormalities. Because of the placental deficiencies, the role of SENP2 in development of the embryonic tissues has not been properly determined. The brain and heart abnormalities may be secondary to placental insufficiency. Here we have created a new mouse strain permitting conditional inactivation of SENP2. Mice homozygous for germline deletion of the conditional allele exhibit trophoblast defects and embryonic abnormalities resembling the global SENP2 knockout. However, tissue-specific disruptions of SENP2 demonstrate its dispensable role in embryogenesis. Placental expression of SENP2 is necessary and sufficient for embryonic heart and brain development. Using a protease deficient model, we further demonstrate the requirement of SENP2-dependent SUMO modification in development of all major trophoblast lineages. SENP2 regulates sumoylation of Mdm2 which controls p53 activities critical for G-S transition of mitotic division and endoreduplication in trophoblast proliferation and differentiation, respectively. The differentiation of trophoblasts is also dependent on SENP2-mediated activation of p57Kip2, a CDK-specific inhibitor required for endoreduplication.
Collapse
|
94
|
Abstract
SUMO posttranslational modification directs gene transcription and epigenetic programming to support normal cell function. The dynamic nature of SUMO-modification makes it difficult to identify endogenous protein substrates. Isolation of chromatin-bound SUMO targets is exceptionally challenging, as conventional immunoprecipitation assays are inefficient at concentrating this protein population. This chapter describes a protocol that effectively precipitates chromatin-associated fractions of SUMOylated heterochromatin protein 1α in cultured cells. Techniques to enrich endogenous SUMO substrates at the chromatin are also demonstrated and discussed. This approach could be adapted to evaluate chromatin-bound SUMO targets in additional in vivo systems.
Collapse
Affiliation(s)
- Tasneem Bawa-Khalfe
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, 3517 Cullen Blvd, SERC Bldg, Rm 3010, Houston, TX, 77204-5056, USA.
| |
Collapse
|
95
|
Wu H, Chen X, Cheng J, Qi Y. SUMOylation and Potassium Channels: Links to Epilepsy and Sudden Death. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 103:295-321. [PMID: 26920693 DOI: 10.1016/bs.apcsb.2015.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuronal potassium ion channels play an essential role in the generation of the action potential and excitability of neurons. The dysfunction of ion channel subunits can cause channelopathies, which are associated in some cases with sudden unexplained death in epilepsy SUDEP. The physiological roles of neuronal ion channels have been largely determined, but little is known about the molecular mechanisms underlying neurological channelopathies, especially the determinants of the channels' regulation. SUMO (small ubiquitin-like modifier) proteins covalently conjugate lysine residues in a large number of target proteins and modify their functions. SUMO modification (SUMOylation) has emerged as an important regulatory mechanism for protein stability, function, subcellular localization, and protein-protein interactions. Since SUMO was discovered almost 20 years ago, the biological contribution of SUMOylation has not fully understood. It is until recently that the physiological impacts of SUMOylation on the regulation of neuronal potassium ion channels have been investigated. It is well established that SUMOylation controls many aspects of nuclear function, but it is now clear that it is also a key determinant in the function of potassium channels, and SUMOylation has also been implicated in a wide range of channelopathies, including epilepsy and sudden death.
Collapse
Affiliation(s)
- Hongmei Wu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, PR China
| | - Xu Chen
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, PR China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yitao Qi
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
96
|
Putiri EL, Tiedemann RL, Liu C, Choi JH, Robertson KD. Impact of human MLL/COMPASS and polycomb complexes on the DNA methylome. Oncotarget 2015; 5:6338-52. [PMID: 25071008 PMCID: PMC4171634 DOI: 10.18632/oncotarget.2215] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The correlation between DNA methylation and a subset of histone post-translational modifications (positive and negative) has hinted at an underlying regulatory crosstalk between histone marks and DNA methylation in patterning the human DNA methylome, an idea further supported by corresponding alterations to both histone marks and DNA methylation during malignant transformation. This study investigated the framework by which histone marks influence DNA methylation at a genome-wide level. Using RNAi in a pluripotent human embryonic carcinoma cell line we depleted essential components of the MLL/COMPASS, polycomb repressive complex 2 (PRC2), and PRC1 histone modifying complexes that establish, respectively, the post-translational modifications H3K4me3, H3K27me3, and H2AK119ub, and assayed the impact of the subsequent depletion of these marks on the DNA methylome. Absence of H2AK119ub resulted predominantly in hypomethylation across the genome. Depletion of H3K4me3 and, surprisingly, H3K27me3 caused CpG island hypermethylation at a subset of loci. Intriguingly, many promoters were co-regulated by all three histone marks, becoming hypermethylated with loss of H3K4me3 or H3K27me3 and hypomethylated with depletion of H2AK119ub, and many of these co-regulated loci were among those commonly targeted for aberrant hypermethylation in cancer. Taken together, our results elucidate novel roles for polycomb and MLL/COMPASS in regulating DNA methylation and define targets of this regulation.
Collapse
Affiliation(s)
- Emily L Putiri
- Department of Molecular Pharmacology and Experimental Therapeutics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Rochelle L Tiedemann
- Department of Molecular Pharmacology and Experimental Therapeutics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN; Cancer Center, Georgia Regents University, Augusta, GA
| | - Chunsheng Liu
- Department of Molecular Pharmacology and Experimental Therapeutics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Jeong-Hyeon Choi
- Department of Molecular Pharmacology and Experimental Therapeutics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN; Cancer Center, Georgia Regents University, Augusta, GA
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics and Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
97
|
Suzawa M, Miranda DA, Ramos KA, Ang KKH, Faivre EJ, Wilson CG, Caboni L, Arkin MR, Kim YS, Fletterick RJ, Diaz A, Schneekloth JS, Ingraham HA. A gene-expression screen identifies a non-toxic sumoylation inhibitor that mimics SUMO-less human LRH-1 in liver. eLife 2015; 4. [PMID: 26653140 PMCID: PMC4749390 DOI: 10.7554/elife.09003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023] Open
Abstract
SUMO-modification of nuclear proteins has profound effects on gene expression. However, non-toxic chemical tools that modulate sumoylation in cells are lacking. Here, to identify small molecule sumoylation inhibitors we developed a cell-based screen that focused on the well-sumoylated substrate, human Liver Receptor Homolog-1 (hLRH-1, NR5A2). Our primary gene-expression screen assayed two SUMO-sensitive transcripts, APOC3 and MUC1, that are upregulated by SUMO-less hLRH-1 or by siUBC9 knockdown, respectively. A polyphenol, tannic acid (TA) emerged as a potent sumoylation inhibitor in vitro (IC50 = 12.8 µM) and in cells. TA also increased hLRH-1 occupancy on SUMO-sensitive transcripts. Most significantly, when tested in humanized mouse primary hepatocytes, TA inhibits hLRH-1 sumoylation and induces SUMO-sensitive genes, thereby recapitulating the effects of expressing SUMO-less hLRH-1 in mouse liver. Our findings underscore the benefits of phenotypic screening for targeting post-translational modifications, and illustrate the potential utility of TA for probing the cellular consequences of sumoylation. DOI:http://dx.doi.org/10.7554/eLife.09003.001 Proteins in cells carry out diverse tasks. One way in which this diversity is achieved by proteins is through the attachment of molecular tags. SUMO is one such tag that can reversibly attach to proteins and alter their activity. The modification of proteins by SUMO is known as sumoylation, and it regulates many processes that are essential for living cells. In particular, transcription factors—the proteins that bind to DNA to switch genes on or off—are highly modified by SUMO. However, the consequences of sumoylation are not fully understood, and current research into this area has been hindered by a lack of effective and non-toxic chemicals that stop or slow down sumoylation. Suzawa, Miranda, Ramos et al. have now screened a large collection of compounds, which had already been approved for medical use, to find one that could inhibit sumoylation without toxic effects. The compounds were tested for their ability to alter the activity of a transcription factor called human Liver Receptor Homolog-1. This protein, which is referred to as LRH-1 for short, is an ideal candidate to test SUMO inhibitors because it is highly modified by multiple SUMO tags. This screen identified a compound from plants called tannic acid as a non-toxic and potent inhibitor of sumoylation. Further experiments confirmed that tannic acid prevented the modification of LHR-1 as well a number of different proteins that also commonly modified by SUMO. Inhibiting the sumoylation of LRH-1 led to an increase in the expression of genes that are normally silenced by SUMO-modified LRH-1. Similar results were obtained when tannic acid was tested using human cells and “humanized” liver cells from mice that had been engineered to express human LRH-1. The next big challenge is to find new chemical probes that can be used to specifically promote or inhibit SUMO modification of just one particular protein. DOI:http://dx.doi.org/10.7554/eLife.09003.002
Collapse
Affiliation(s)
- Miyuki Suzawa
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Diego A Miranda
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Karmela A Ramos
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Kenny K-H Ang
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Emily J Faivre
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Christopher G Wilson
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Laura Caboni
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Michelle R Arkin
- Small Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Yeong-Sang Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, United States
| | - Robert J Fletterick
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Aaron Diaz
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, United States
| | - John S Schneekloth
- Chemical Biology Laboratory, National Cancer Institute, Frederick, United States
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
98
|
Domingues P, Golebiowski F, Tatham MH, Lopes AM, Taggart A, Hay RT, Hale BG. Global Reprogramming of Host SUMOylation during Influenza Virus Infection. Cell Rep 2015; 13:1467-1480. [PMID: 26549460 PMCID: PMC4660286 DOI: 10.1016/j.celrep.2015.10.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/24/2015] [Accepted: 09/28/2015] [Indexed: 12/22/2022] Open
Abstract
Dynamic nuclear SUMO modifications play essential roles in orchestrating cellular responses to proteotoxic stress, DNA damage, and DNA virus infection. Here, we describe a non-canonical host SUMOylation response to the nuclear-replicating RNA pathogen, influenza virus, and identify viral RNA polymerase activity as a major contributor to SUMO proteome remodeling. Using quantitative proteomics to compare stress-induced SUMOylation responses, we reveal that influenza virus infection triggers unique re-targeting of SUMO to 63 host proteins involved in transcription, mRNA processing, RNA quality control, and DNA damage repair. This is paralleled by widespread host deSUMOylation. Depletion screening identified ten virus-induced SUMO targets as potential antiviral factors, including C18orf25 and the SMC5/6 and PAF1 complexes. Mechanistic studies further uncovered a role for SUMOylation of the PAF1 complex component, parafibromin (CDC73), in potentiating antiviral gene expression. Our global characterization of influenza virus-triggered SUMO redistribution provides a proteomic resource to understand host nuclear SUMOylation responses to infection.
Collapse
Affiliation(s)
- Patricia Domingues
- MRC-University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Filip Golebiowski
- MRC-University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Michael H Tatham
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Antonio M Lopes
- MRC-University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Aislynn Taggart
- MRC-University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Ronald T Hay
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Benjamin G Hale
- MRC-University of Glasgow Centre for Virus Research, Garscube Campus, 464 Bearsden Road, Glasgow G61 1QH, UK.
| |
Collapse
|
99
|
Sowpati DT, Ramamoorthy S, Mishra RK. Expansion of the polycomb system and evolution of complexity. Mech Dev 2015; 138 Pt 2:97-112. [DOI: 10.1016/j.mod.2015.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 11/28/2022]
|
100
|
Evaluation of the activity and substrate specificity of the human SENP family of SUMO proteases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:139-47. [PMID: 26522917 DOI: 10.1016/j.bbamcr.2015.10.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/05/2015] [Accepted: 10/28/2015] [Indexed: 11/24/2022]
Abstract
Protein modification with the small ubiquitin-like modifier (SUMO) is a reversible process regulating many central biological pathways. The reversibility of SUMOylation is ensured by SUMO proteases many of which belong to the sentrin/SUMO-specific protease (SENP) family. In recent years, many advances have been made in allocating SENPs to specific biological pathways. However, due to difficulties in obtaining recombinant full-length active SENPs for thorough enzymatic characterization, our knowledge on these proteases is still limited. In this work, we used in vitro synthesized full-length human SENPs to perform a side-by-side comparison of their activities and substrate specificities. ProSUMO1/2/3, RanGAP1-SUMO1/2/3 and polySUMO2/3 chains were used as substrates in these analyses. We found that SENP1 is by far the most versatile and active SENP whereas SENP3 stands out as the least active of these enzymes. Finally, a comparison between the activities of full-length SENPs and their catalytic domains suggests that in some cases their non-catalytic regions influence their activity.
Collapse
|