1
|
Rashpa R, Klages N, Schvartz D, Pasquarello C, Brochet M. The Skp1-Cullin1-FBXO1 complex is a pleiotropic regulator required for the formation of gametes and motile forms in Plasmodium berghei. Nat Commun 2023; 14:1312. [PMID: 36898988 PMCID: PMC10006092 DOI: 10.1038/s41467-023-36999-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Malaria-causing parasites of the Plasmodium genus undergo multiple developmental phases in the human and the mosquito hosts, regulated by various post-translational modifications. While ubiquitination by multi-component E3 ligases is key to regulate a wide range of cellular processes in eukaryotes, little is known about its role in Plasmodium. Here we show that Plasmodium berghei expresses a conserved SKP1/Cullin1/FBXO1 (SCFFBXO1) complex showing tightly regulated expression and localisation across multiple developmental stages. It is key to cell division for nuclear segregation during schizogony and centrosome partitioning during microgametogenesis. It is additionally required for parasite-specific processes including gamete egress from the host erythrocyte, as well as integrity of the apical and the inner membrane complexes (IMC) in merozoite and ookinete, two structures essential for the dissemination of these motile stages. Ubiquitinomic surveys reveal a large set of proteins ubiquitinated in a FBXO1-dependent manner including proteins important for egress and IMC organisation. We additionally demonstrate an interplay between FBXO1-dependent ubiquitination and phosphorylation via calcium-dependent protein kinase 1. Altogether we show that Plasmodium SCFFBXO1 plays conserved roles in cell division and is also important for parasite-specific processes in the mammalian and mosquito hosts.
Collapse
Affiliation(s)
- Ravish Rashpa
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland.
| | - Natacha Klages
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland
| | - Domitille Schvartz
- University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Carla Pasquarello
- University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Mathieu Brochet
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland.
| |
Collapse
|
2
|
Non-proteolytic ubiquitylation in cellular signaling and human disease. Commun Biol 2022; 5:114. [PMID: 35136173 PMCID: PMC8826416 DOI: 10.1038/s42003-022-03060-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022] Open
Abstract
Ubiquitylation is one of the most common post-translational modifications (PTMs) of proteins that frequently targets substrates for proteasomal degradation. However it can also result in non-proteolytic events which play important functions in cellular processes such as intracellular signaling, membrane trafficking, DNA repair and cell cycle. Emerging evidence demonstrates that dysfunction of non-proteolytic ubiquitylation is associated with the development of multiple human diseases. In this review, we summarize the current knowledge and the latest concepts on how non-proteolytic ubiquitylation pathways are involved in cellular signaling and in disease-mediating processes. Our review, may advance our understanding of the non-degradative ubiquitylation process. Evanthia Pangou and co-authors review recent insights into the important roles of non-proteolytic ubiquitylation in cellular signaling as well as in physiology and disease.
Collapse
|
3
|
Piedade WP, Famulski JK. E3 ubiquitin ligase-mediated regulation of vertebrate ocular development; new insights into the function of SIAH enzymes. Biochem Soc Trans 2021; 49:327-340. [PMID: 33616626 PMCID: PMC7924998 DOI: 10.1042/bst20200613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/18/2023]
Abstract
Developmental regulation of the vertebrate visual system has been a focus of investigation for generations as understanding this critical time period has direct implications on our understanding of congenital blinding disease. The majority of studies to date have focused on transcriptional regulation mediated by morphogen gradients and signaling pathways. However, recent studies of post translational regulation during ocular development have shed light on the role of the ubiquitin proteasome system (UPS). This rather ubiquitous yet highly diverse system is well known for regulating protein function and localization as well as stability via targeting for degradation by the 26S proteasome. Work from many model organisms has recently identified UPS activity during various milestones of ocular development including retinal morphogenesis, retinal ganglion cell function as well as photoreceptor homeostasis. In particular work from flies and zebrafish has highlighted the role of the E3 ligase enzyme family, Seven in Absentia Homologue (Siah) during these events. In this review, we summarize the current understanding of UPS activity during Drosophila and vertebrate ocular development, with a major focus on recent findings correlating Siah E3 ligase activity with two major developmental stages of vertebrate ocular development, retinal morphogenesis and photoreceptor specification and survival.
Collapse
|
4
|
Jerabkova K, Liao Y, Kleiss C, Fournane S, Durik M, Agote-Arán A, Brino L, Sedlacek R, Sumara I. Deubiquitylase UCHL3 regulates bi-orientation and segregation of chromosomes during mitosis. FASEB J 2020; 34:12751-12767. [PMID: 32738097 DOI: 10.1096/fj.202000769r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/03/2020] [Accepted: 07/14/2020] [Indexed: 11/11/2022]
Abstract
Equal segregation of chromosomes during mitosis ensures euploidy of daughter cells. Defects in this process may result in an imbalance in the chromosomal composition and cellular transformation. Proteolytic and non-proteolytic ubiquitylation pathways ensure directionality and fidelity of mitotic progression but specific mitotic functions of deubiquitylating enzymes (DUBs) remain less studied. Here we describe the role of the DUB ubiquitin carboxyl-terminal hydrolase isozyme L3 (UCHL3) in the regulation of chromosome bi-orientation and segregation during mitosis. Downregulation or inhibition of UCHL3 leads to chromosome alignment defects during metaphase. Frequent segregation errors during anaphase are also observed upon inactivation of UCHL3. Mechanistically, UCHL3 interacts with and deubiquitylates Aurora B, the catalytic subunit of chromosome passenger complex (CPC), known to be critically involved in the regulation of chromosome alignment and segregation. UCHL3 does not regulate protein levels of Aurora B or the binding of Aurora B to other CPC subunits. Instead, UCHL3 promotes localization of Aurora B to kinetochores, suggesting its role in the error correction mechanism monitoring bi-orientation of chromosomes during metaphase. Thus, UCHL3 contributes to the regulation of faithful genome segregation and maintenance of euploidy in human cells.
Collapse
Affiliation(s)
- Katerina Jerabkova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, v.v.i., Vestec, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Yongrong Liao
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Charlotte Kleiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Sadek Fournane
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Matej Durik
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Arantxa Agote-Arán
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Laurent Brino
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, v.v.i., Vestec, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic.,Czech Centre for Phenogenomics, Institute of Molecular Genetics of the CAS, v.v.i., Vestec, Czech Republic
| | - Izabela Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of development and stem cells, Illkirch, France.,Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France.,Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France.,Université de Strasbourg, Strasbourg, France
| |
Collapse
|
5
|
Deubiquitinating Enzymes: A Critical Regulator of Mitosis. Int J Mol Sci 2019; 20:ijms20235997. [PMID: 31795161 PMCID: PMC6929034 DOI: 10.3390/ijms20235997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022] Open
Abstract
Mitosis is a complex and dynamic process that is tightly regulated by a large number of mitotic proteins. Dysregulation of these proteins can generate daughter cells that exhibit genomic instability and aneuploidy, and such cells can transform into tumorigenic cells. Thus, it is important for faithful mitotic progression to regulate mitotic proteins at specific locations in the cells at a given time in each phase of mitosis. Ubiquitin-dependent modifications play critical roles in this process by regulating the degradation, translocation, or signal transduction of mitotic proteins. Here, we review how ubiquitination and deubiquitination regulate the progression of mitosis. In addition, we summarize the substrates and roles of some deubiquitinating enzymes (DUBs) crucial for mitosis and describe how they contribute error correction during mitosis and control the transition between the mitotic phases.
Collapse
|
6
|
Lu G, Yi J, Gubas A, Wang YT, Wu Y, Ren Y, Wu M, Shi Y, Ouyang C, Tan HWS, Wang T, Wang L, Yang ND, Deng S, Xia D, Chen RH, Tooze SA, Shen HM. Suppression of autophagy during mitosis via CUL4-RING ubiquitin ligases-mediated WIPI2 polyubiquitination and proteasomal degradation. Autophagy 2019; 15:1917-1934. [PMID: 30898011 DOI: 10.1080/15548627.2019.1596484] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Macroautophagy/autophagy is a cellular process in which cytosolic contents are degraded by lysosome in response to various stress conditions. Apart from its role in the maintenance of cellular homeostasis, autophagy also involves in regulation of cell cycle progression under nutrient-deprivation conditions. However, whether and how autophagy is regulated by the cell cycle especially during mitosis remains largely undefined. Here we show that WIPI2/ATG18B (WD repeat domain, phosphoinositide interacting 2), an autophagy-related (ATG) protein that plays a critical role in autophagosome biogenesis, is a direct substrate of CUL4-RING ubiquitin ligases (CRL4s). Upon mitosis induction, CRL4s are activated via neddylation, and recruit WIPI2 via DDB1 (damage specific DNA binding protein 1), leading to polyubiquitination and proteasomal degradation of WIPI2 and suppression of autophagy. The WIPI2 protein level and autophagy during mitosis could be rescued by knockdown of CRL4s or treatment with MLN4924/Pevonedistat, a selective inhibitor of CRLs, via suppression of NAE1 (NEDD8 activating enzyme E1 subunit 1). Moreover, restoration of WIPI2 rescues autophagy during mitosis and leads to mitotic slippage and cell senescence. Our study thus discovers a novel function of CRL4s in autophagy by targeting WIPI2 for polyubiquitination and proteasomal degradation during mitosis. Abbreviations: ACTB, actin beta; ATG, autophagy-related; AMPK, AMP-activated protein kinase; AURKB/ARK2, aurora kinase B; BafA1, bafilomycin A1; CCNB1, cyclin B1; CDK1, cyclin dependent kinase 1; CHX, cycloheximide; CQ, chloroquine; CRL4s, CUL4-RING ubiquitin ligases; DDB1, damage specific DNA binding protein 1; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; GFP, green fluorescent protein; GST, glutathione S-transferase; MAP1LC3B/LC3B, microtubule associated protein 1 light chain 3 beta; STK11/LKB1,serine/threonine kinase 11; MTORC1/MTOR complex 1, mechanistic target of rapamycin kinase complex 1; NAE1, NEDD8 activating enzyme E1 subunit 1; NOC, nocodazole; RING, really interesting new gene; RBX1, ring-box 1; SA-GLB1/β-gal, senescence-associated galactosidase beta 1; TSC2, TSC complex subunit 2; TUBA, tubulin alpha; WIPI2, WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Guang Lu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Juan Yi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Andrea Gubas
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute , London UK
| | - Ya-Ting Wang
- Institute of Biological Chemistry, Academia Sinica , Taipei , Taiwan
| | - Yihua Wu
- School of Public Health, Zhejiang University , Hangzhou , China
| | - Yi Ren
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Man Wu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore.,School of Pharmacy, Shanghai University of Traditional Chinese Medicine , Shanghai , China
| | - Yin Shi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore.,Department of Biochemistry and Molecular Biology, Zhejiang University School of Medicine , Hangzhou , China
| | - Chenxi Ouyang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Hayden Weng Siong Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore , Singapore
| | - Tianru Wang
- Life Sciences Program, Faculty of Arts and Sciences, University of Toronto , Toronto , Canada
| | - Liming Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Nai-Di Yang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Dajing Xia
- School of Public Health, Zhejiang University , Hangzhou , China
| | - Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica , Taipei , Taiwan
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute , London UK
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore , Singapore
| |
Collapse
|
7
|
Meza Gutierrez F, Simsek D, Mizrak A, Deutschbauer A, Braberg H, Johnson J, Xu J, Shales M, Nguyen M, Tamse-Kuehn R, Palm C, Steinmetz LM, Krogan NJ, Toczyski DP. Genetic analysis reveals functions of atypical polyubiquitin chains. eLife 2018; 7:42955. [PMID: 30547882 PMCID: PMC6305200 DOI: 10.7554/elife.42955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/30/2018] [Indexed: 12/27/2022] Open
Abstract
Although polyubiquitin chains linked through all lysines of ubiquitin exist, specific functions are well-established only for lysine-48 and lysine-63 linkages in Saccharomyces cerevisiae. To uncover pathways regulated by distinct linkages, genetic interactions between a gene deletion library and a panel of lysine-to-arginine ubiquitin mutants were systematically identified. The K11R mutant had strong genetic interactions with threonine biosynthetic genes. Consistently, we found that K11R mutants import threonine poorly. The K11R mutant also exhibited a strong genetic interaction with a subunit of the anaphase-promoting complex (APC), suggesting a role in cell cycle regulation. K11-linkages are important for vertebrate APC function, but this was not previously described in yeast. We show that the yeast APC also modifies substrates with K11-linkages in vitro, and that those chains contribute to normal APC-substrate turnover in vivo. This study reveals comprehensive genetic interactomes of polyubiquitin chains and characterizes the role of K11-chains in two biological pathways.
Collapse
Affiliation(s)
- Fernando Meza Gutierrez
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | | | - Arda Mizrak
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | | | - Hannes Braberg
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Jeffrey Johnson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Jiewei Xu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Michael Shales
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - Michelle Nguyen
- Stanford Genome Technology Center, Stanford University, Stanford, United States
| | - Raquel Tamse-Kuehn
- Stanford Genome Technology Center, Stanford University, Stanford, United States
| | - Curt Palm
- Stanford Genome Technology Center, Stanford University, Stanford, United States
| | - Lars M Steinmetz
- Stanford Genome Technology Center, Stanford University, Stanford, United States
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States
| | - David P Toczyski
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
8
|
Yu H, Xu W, Gong F, Chi B, Chen J, Zhou L. MicroRNA-155 regulates the proliferation, cell cycle, apoptosis and migration of colon cancer cells and targets CBL. Exp Ther Med 2017; 14:4053-4060. [PMID: 29104623 PMCID: PMC5658697 DOI: 10.3892/etm.2017.5085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 06/08/2017] [Indexed: 12/26/2022] Open
Abstract
MicroRNA-155 (miR-155) is a well-studied miR and acts as an oncomiR in numerous cancer types. However, the biological functions of miR-155 in colon cancer as well as its target genes have remained to be fully elucidated. In order to investigate the biological functions of miR-155, MTT, colony formation and wound healing assays, cell cycle analysis and detection of apoptosis were performed. The results demonstrated that miR-155 promoted the proliferation of colon cancer cells and enhanced their colony formation capacity, promoted their cell cycle progression and inhibited apoptosis. miR-155 also promoted the migration of colon cancer cells. In the present study, casitas B-lineage lymphoma was identified as a novel target of miR-155. The present study suggested that miR-155 functions as an oncomiR in colon cancer cells and may become a promising therapeutic target for colon cancer therapy.
Collapse
Affiliation(s)
- Hua Yu
- Department of General Surgery, The Branch of Shanghai First People's Hospital, Shanghai 200081, P.R. China
| | - Weiling Xu
- Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China.,Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fangchao Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Baorong Chi
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Junyi Chen
- Department of General Surgery, The Branch of Shanghai First People's Hospital, Shanghai 200081, P.R. China
| | - Ling Zhou
- Department of General Surgery, The Branch of Shanghai First People's Hospital, Shanghai 200081, P.R. China
| |
Collapse
|
9
|
Huang G, Kaufman AJ, Xu K, Manova K, Singh B. Squamous cell carcinoma-related oncogene (SCCRO) neddylates Cul3 protein to selectively promote midbody localization and activity of Cul3 KLHL21 protein complex during abscission. J Biol Chem 2017; 292:15254-15265. [PMID: 28620047 DOI: 10.1074/jbc.m117.778530] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/30/2017] [Indexed: 12/16/2022] Open
Abstract
Squamous cell carcinoma-related oncogene (SCCRO)/DCUN1D1, a component of the neddylation E3 complex, regulates the activity of the cullin-RING-ligase type of ubiquitination E3s by promoting neddylation of cullin family members. Studies have shown that SCCRO regulates proliferation in vitro and in vivo Here we show that inactivation of SCCRO results in prolonged mitotic time because of delayed and/or failed abscission. The effects of SCCRO on abscission involve its role in neddylation and localization of Cul3 to the midbody. The Cul3 adaptor KLHL21 mediates the effects of SCCRO on abscission, as it fails to localize to the midbody in SCCRO-deficient cells during abscission, and its inactivation resulted in phenotypic changes identical to SCCRO inactivation. Ubiquitination-promoted turnover of Aurora B at the midbody was deficient in SCCRO- and KLHL21-deficient cells, suggesting that it is the target of Cul3KLHL21 at the midbody. Correction of abscission delays in SCCRO-deficient cells with addition of an Aurora B inhibitor at the midbody stage suggests that Aurora B is the target of SCCRO-promoted Cul3KLHL21 activity. The activity of other Cul3-anchored complexes, including Cul3KLHL9/KLHL13, was intact in SCCRO-deficient cells, suggesting that SCCRO selectively, rather than collectively, neddylates cullins in vivo Combined, these findings support a model in which the SCCRO, substrate, and substrate adaptors cooperatively provide tight control of neddylation and cullin-RING-ligase activity in vivo.
Collapse
Affiliation(s)
- Guochang Huang
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology and
| | - Andrew J Kaufman
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology and
| | - Ke Xu
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Katia Manova
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Bhuvanesh Singh
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology and
| |
Collapse
|
10
|
Hatano Y, Naoki K, Suzuki A, Ushimaru T. Positive feedback promotes mitotic exit via the APC/C-Cdh1-separase-Cdc14 axis in budding yeast. Cell Signal 2016; 28:1545-54. [DOI: 10.1016/j.cellsig.2016.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 06/30/2016] [Accepted: 07/08/2016] [Indexed: 10/21/2022]
|
11
|
Zhao Y, Yang X. Regulation of sensitivity of tumor cells to antitubulin drugs by Cdk1-TAZ signalling. Oncotarget 2016; 6:21906-17. [PMID: 26183396 PMCID: PMC4673135 DOI: 10.18632/oncotarget.4259] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/22/2015] [Indexed: 12/22/2022] Open
Abstract
Antitubulin drugs are commonly used for the treatment of numerous cancers. However, either the intrinsic or acquired resistances of patients to these drugs result in the failure of the treatment and high mortality of cancers. Therefore, identifying genes or signalling pathways involved in antitubulin drug resistances is critical for future successful treatment of cancers. TAZ (Transcriptional coactivator with PDZ-binding motif), which is a core component of the Hippo pathway, is overexpressed in various cancers. We have recently shown that high levels of TAZ in cancer cells result in Taxol resistance through up-regulation of downstream targets Cyr61 and CTGF. However, how TAZ is regulated in response to Taxol is largely unknown. In this study, we found that Cdk1 (Cyclin-dependent kinase 1) directly phosphorylated TAZ on six novel sites independent of the Hippo pathway, which further resulted in TAZ degradation through proteasome system. Phosphorylation-mimicking TAZ mutant was unstable, and therefore abolished TAZ-induced antitubulin drug resistances. This study provides first evidence that Cdk1 is a novel kinase phosphorylating and regulating TAZ stability and suggests that Cdk1-TAZ signalling is a critical regulator of antitubulin drug response in cancer cells and may be a potential target for the treatment of antitubulin-drug resistant cancer patients.
Collapse
Affiliation(s)
- Yulei Zhao
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L3N6, Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L3N6, Canada
| |
Collapse
|
12
|
França JA, Diniz MG, Bernardes VF, Costa-Silva RC, Souza RP, Gomez RS, Gomes CC. Cohesin subunits, STAG1 and STAG2, and cohesin regulatory factor, PDS5b, in oral squamous cells carcinomas. J Oral Pathol Med 2016; 46:188-193. [PMID: 27341316 DOI: 10.1111/jop.12474] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cohesin complex is responsible for sister chromatid cohesion. STAG1/STAG2 is part of the complex, which is regulated by PDS5B. Alterations in these genes were described in tumors. PDS5B is a negative regulator of cell proliferation. We aimed to assess molecular alterations in these genes in oral squamous cell carcinoma (OSCC) and predict their expression by the expression of 84 cell cycle genes. In addition, we investigated whether pds5b protein expression impacted ki-67 and p53 immunopositivity. METHODS We assessed loss of heterozygosity (LOH) at STAG1 and STAG2 loci in 15 OSCC using three polymorphic markers. Associations between the immunoexpression of pds5b and ki-67 and p53 were tested in 62 samples. Differences between transcriptional levels of STAG1, STAG2, and PDS5B between OSCC and normal oral mucosa (NM) were evaluated by qPCR. An 84 cell cycle genes qPCR array was carried with OSCC samples, and STAG1, STAG2, and PDS5B were independently used as response variables in multiple linear regression models. RESULTS Loss of heterozygosity in at least one marker was observed in three samples. pds5b, p53, and ki-67 were highly expressed, and no association was found between pds5b immunoexpression and ki-67 or p53 (P > 0.05). OSCC and NM showed similar transcriptional levels of STAG1, STAG2, and PDS5B. STAG1 and CUL3 expression seem to be related (P = 0.004). CONCLUSIONS There is LOH at STAG1 and STAG2 loci in OSCC, but OSCC and NM showed similar transcriptional levels of STAG1, STAG2, and PDS5B. pds5b immunoexpression in OSCC was high, but it was not associated with proliferation cell index.
Collapse
Affiliation(s)
- Josiane Alves França
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| | - Marina Gonçalves Diniz
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| | - Vanessa Fátima Bernardes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| | - Raíssa Cristina Costa-Silva
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| | - Renan Pedra Souza
- Department of General Biology, Biological Sciences Institute, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| | - Carolina Cavaliéri Gomes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte, Brazil
| |
Collapse
|
13
|
Late mitotic functions of Aurora kinases. Chromosoma 2016; 126:93-103. [DOI: 10.1007/s00412-016-0594-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
|
14
|
Abstract
WWP2 is a ubiquitin E3 ligase belonging to the Nedd4-like family. Given that WWP2 target proteins including PTEN that are crucial for regulating cell proliferation or suppressing tumorigenesis, we have asked whether WWP2 plays a role in controlling cell cycle progression. Here we report that WWP2 is necessary for normal cell cycle progression as its silencing significantly reduces the cell proliferation rate. We have identified that an isoform of WWP2 (WWP2-V4) is highly expressed in the M phase of the cell cycle. Silencing of WWP2 accelerates the turnover of cyclin E, which is accompanied by increased levels of phospho-histone H3 (p-H3) and cyclin B. Moreover, silencing of WWP2 results in compromised phosphorylation of Akt(S473), a residue whose phosphorylation is tightly associated with the activation of the kinase. Combined, these results strongly suggest that WWP2 is an important component in regulating the Akt signaling cascade, as well as cell cycle progression.
Collapse
Affiliation(s)
- Byeong Hyeok Choi
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo Park, NY, USA
| | - Xun Che
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo Park, NY, USA
| | - Changyan Chen
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Luo Lu
- Division of Molecular Medicine, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Torrance, CA, USA
| | - Wei Dai
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo Park, NY, USA
| |
Collapse
|
15
|
Keum YS, Choi BY. Molecular and chemical regulation of the Keap1-Nrf2 signaling pathway. Molecules 2014; 19:10074-89. [PMID: 25014534 PMCID: PMC6270911 DOI: 10.3390/molecules190710074] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 06/02/2014] [Accepted: 07/02/2014] [Indexed: 12/30/2022] Open
Abstract
Extracellular and intracellular oxidants or electrophiles are key contributors to the damages in cellular macromolecules, such as DNA, proteins and lipids. Nrf2 is a master transcription factor that modulates a cellular antioxidant response program and plays an important role in the protection against oxidants and electrophiles. Keap1 is a regulator of Nrf2 by serving as a substrate adaptor for Cullin3-dependent E3 ubiquitin ligase. While Nrf2 activation is a feasible strategy for treatment of age-related diseases, aberrant Nrf2 activation also confers a selective growth advantage of tumor cells during chemotherapy or radiotherapy. In the present review, we provide an overview of the Keap1-Nrf2-ARE system, the domain organization of Nrf2 and Keap1, and the regulatory mechanisms of Nrf2 proteolysis by Keap1. We also discuss how Nrf2 prevents tumor promotion, hampers the sensitivity of selected tumors against chemotherapy or radiotherapy, and reprograms the metabolism to facilitate the tumor proliferation. Finally, we illustrate the current status in the development of Nrf2 chemical activators and inhibitors for the use of potential chemopreventive agents and chemotherapeutic adjuvants, respectively.
Collapse
Affiliation(s)
- Young-Sam Keum
- College of Pharmacy, Dongguk University, 813-4 Siksa-dong, Goyang, Gyeonggi-do 410-820, Korea.
| | - Bu Young Choi
- Department of Pharmaceutical Science and Engineering, Seowon University, Cheongju, Chungbuk 361-742, Korea.
| |
Collapse
|
16
|
Keum YS. Regulation of Nrf2-Mediated Phase II Detoxification and Anti-oxidant Genes. Biomol Ther (Seoul) 2014; 20:144-51. [PMID: 24116287 PMCID: PMC3792210 DOI: 10.4062/biomolther.2012.20.2.144] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 07/30/2011] [Accepted: 08/17/2011] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanisms by which a variety of naturally-occurring dietary compounds exert chemopreventive effects have been a subject of intense scientific investigations. Induction of phase II detoxification and anti-oxidant enzymes through activation of Nrf2/ARE-dependent gene is recognized as one of the major cellular defense mechanisms against oxidative or xenobiotic stresses and currently represents a critical chemopreventive mechanism of action. In the present review, the functional significance of Keap1/Nrf2 protein module in regulating ARE-dependent phase II detoxification and anti-oxidant gene expression is discussed. The biochemical mechanisms underlying the phosphorylation and expression of Keap1/Nrf2 proteins that are controlled by the intracellular signaling kinases and ubiquitin-mediated E3 ligase system as well as control of nucleocytoplasmic translocation of Nrf2 by its innate nuclear export signal (NES) are described.
Collapse
Affiliation(s)
- Young-Sam Keum
- Department of Biochemistry, College of Pharmacy, Dongguk University, Goyang 410-773, Republic of Korea
| |
Collapse
|
17
|
Ma L, Aslanian A, Sun H, Jin M, Shi Y, Yates JR, Hunter T. Identification of small ubiquitin-like modifier substrates with diverse functions using the Xenopus egg extract system. Mol Cell Proteomics 2014; 13:1659-75. [PMID: 24797264 DOI: 10.1074/mcp.m113.035626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Post-translational modification by SUMO is a highly conserved pathway in eukaryotes that plays very important regulatory roles in many cellular processes. Deregulation of the SUMO pathway contributes to the development and progression of many diseases including cancer. Therefore, identifying additional SUMO substrates and studying how their cellular and biological functions are regulated by sumoylation should provide new insights. Our studies showed that sumoylation activity was significant in Xenopus egg extracts, and that a high level of sumoylation was associated with sperm chromatin when SUMO was incubated with Xenopus egg extracts. By isolating SUMO-conjugated substrates using His-tagged SUMO1 or SUMO2 proteins under denaturing conditions, we identified 346 proteins by mass spectrometry analysis that were not present in control pull-downs. Among them, 167 proteins were identified from interphase egg extracts, 86 proteins from mitotic phase egg extracts, and 93 proteins from both. Thirty-three proteins were pulled down by SUMO1, 85 proteins by SUMO2, and 228 proteins by both. We validated the sumoylation of five candidates, CKB, ATXN10, BTF3, HABP4, and BZW1, by co-transfecting them along with SUMO in HEK293T cells. Gene ontology analysis showed that SUMO substrates identified in this study were involved in diverse biological processes. Additionally, SUMO substrates identified from different cell cycle stages or pulled down by different SUMO homologs were enriched for distinct cellular components and functional categories. Our results comprehensively profile the sumoylation occurring in the Xenopus egg extract system.
Collapse
Affiliation(s)
- Li Ma
- From the ‡Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Aaron Aslanian
- From the ‡Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA; §Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Huaiyu Sun
- From the ‡Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Mingji Jin
- From the ‡Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Yu Shi
- From the ‡Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - John R Yates
- §Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Tony Hunter
- From the ‡Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| |
Collapse
|
18
|
Regulation of mIκBNS stability through PEST-mediated degradation by proteasome. Biochem Biophys Res Commun 2014; 443:1291-5. [PMID: 24406168 DOI: 10.1016/j.bbrc.2013.12.140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 12/29/2013] [Indexed: 12/15/2022]
Abstract
Negative regulatory proteins in a cytokine signaling play a critical role in restricting unwanted excess activation of the signaling pathway. At the same time, negative regulatory proteins need to be removed rapidly from cells to respond properly to the next incoming signal. A nuclear IκB protein called IκBNS is known to inhibit a subset of NF-κB target genes upon its expression by NF-κB activation. Here, we show a mechanism to control the stability of mIκBNS which might be important for cells to prepare the next round signaling. We found that mIκBNS is a short-lived protein of which the stability is controlled by proteasome, independent of ubiquitylation process. We identified that the N-terminal PEST sequence in mIκBNS was critical for the regulation of stability.
Collapse
|
19
|
Kim IS, Lee M, Park JH, Jeon R, Baek SH, Kim KI. βTrCP-mediated ubiquitylation regulates protein stability of Mis18β in a cell cycle-dependent manner. Biochem Biophys Res Commun 2014; 443:62-7. [DOI: 10.1016/j.bbrc.2013.11.058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 11/12/2013] [Indexed: 10/26/2022]
|
20
|
Leljak-Levanić D, Juranić M, Sprunck S. De novo zygotic transcription in wheat (Triticum aestivum L.) includes genes encoding small putative secreted peptides and a protein involved in proteasomal degradation. PLANT REPRODUCTION 2013; 26:267-85. [PMID: 23912470 DOI: 10.1007/s00497-013-0229-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/10/2013] [Indexed: 05/12/2023]
Abstract
Wheat is one of the world's most important crops, and increasing grain yield is a major challenge for the future. Still, our knowledge about the molecular machineries responsible for early post-fertilization events such as zygotic reprogramming, the initial cell-specification events during embryogenesis, and the intercellular communication between the early embryo and the developing endosperm is very limited. Here, we describe the identification of de novo transcribed genes in the wheat zygote. We used wheat ovaries of defined post-fertilization stages to isolate zygotes and early embryos, and identified genes that are specifically induced in these particular stages. Importantly, we observed that some of the zygotic-induced genes encode proteins with similarity to secreted signaling peptides such as TAPETUM DETERMINANT 1 and EGG APPARATUS 1, and to MATH-BTB proteins which are known substrate-binding adaptors for the Cullin3-based ubiquitin E3 ligase. This suggests that both cell-cell signaling and targeted proteasomal degradation may be important molecular events during zygote formation and the progression of early embryogenesis.
Collapse
Affiliation(s)
- Dunja Leljak-Levanić
- Department of Molecular Biology, Faculty of Science and Mathematics, University of Zagreb, Horvatovac 102a, 10000, Zagreb, Croatia
| | | | | |
Collapse
|
21
|
Metzger T, Kleiss C, Sumara I. CUL3 and protein kinases: insights from PLK1/KLHL22 interaction. Cell Cycle 2013; 12:2291-6. [PMID: 24067371 PMCID: PMC3755079 DOI: 10.4161/cc.25369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
Posttranslational mechanisms drive fidelity of cellular processes. Phosphorylation and ubiquitination of substrates represent very common, covalent, posttranslational modifications and are often co-regulated. Phosphorylation may play a critical role both by directly regulating E3-ubiquitin ligases and/or by ensuring specificity of the ubiquitination substrate. Importantly, many kinases are not only critical regulatory components of these pathways but also represent themselves the direct ubiquitination substrates. Recent data suggest the role of CUL3-based ligases in both proteolytic and non-proteolytic regulation of protein kinases. Our own recent study identified the mitotic kinase PLK1 as a direct target of the CUL3 E3-ligase complex containing BTB-KELCH adaptor protein KLHL22. (1) In this study, we aim at gaining mechanistic insights into CUL3-mediated regulation of the substrates, in particular protein kinases, by analyzing mechanisms of interaction between KLHL22 and PLK1. We find that kinase activity of PLK1 is redundant for its targeting for CUL3-ubiquitination. Moreover, CUL3/KLHL22 may contact 2 distinct motifs within PLK1 protein, consistent with the bivalent mode of substrate targeting found in other CUL3-based complexes. We discuss these findings in the context of the existing knowledge on other protein kinases and substrates targeted by CUL3-based E3-ligases.
Collapse
Affiliation(s)
- Thibaud Metzger
- Institute of Genetics and Molecular and Cellular Biology (IGBMC); Illkirch, France
| | - Charlotte Kleiss
- Institute of Genetics and Molecular and Cellular Biology (IGBMC); Illkirch, France
| | - Izabela Sumara
- Institute of Genetics and Molecular and Cellular Biology (IGBMC); Illkirch, France
| |
Collapse
|
22
|
Abstract
Conjugation of ubiquitin (ubiquitination) to substrate proteins is a widespread modification that ensures fidelity of many cellular processes. During mitosis, different dynamic morphological transitions have to be coordinated in a temporal and spatial manner to allow for precise partitioning of the genetic material into two daughter cells, and ubiquitination of key mitotic factors is believed to provide both directionality and fidelity to this process. While directionality can be achieved by a proteolytic type of ubiquitination signal, the fidelity is often determined by various types of ubiquitin conjugation that does not target substrates for proteolysis by the proteasome. An additional level of complexity is provided by various ubiquitin-interacting proteins that act downstream of the ubiquitinated substrate and can serve as "decoders" for the ubiquitin signal. They may, specifically reverse ubiquitin attachment (deubiquitinating enzymes, DUBs) or, act as a receptor for transfer of the ubiquitinated substrate toward downstream signaling components and/or subcellular compartments (ubiquitin-binding proteins, UBPs). In this review, we aim at summarizing the knowledge and emerging concepts about the role of ubiquitin decoders, DUBs, and UBPs that contribute to faithful regulation of mitotic division.
Collapse
Affiliation(s)
- Sadek Fournane
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
| | | | | | | |
Collapse
|
23
|
Beck J, Maerki S, Posch M, Metzger T, Persaud A, Scheel H, Hofmann K, Rotin D, Pedrioli P, Swedlow JR, Peter M, Sumara I. Ubiquitylation-dependent localization of PLK1 in mitosis. Nat Cell Biol 2013; 15:430-9. [PMID: 23455478 PMCID: PMC7116173 DOI: 10.1038/ncb2695] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 01/18/2013] [Indexed: 11/09/2022]
Abstract
Polo-like kinase 1 (PLK1) critically regulates mitosis through its dynamic localization to kinetochores, centrosomes and the midzone. The polo-box domain (PBD) and activity of PLK1 mediate its recruitment to mitotic structures, but the mechanisms regulating PLK1 dynamics remain poorly understood. Here, we identify PLK1 as a target of the cullin 3 (CUL3)-based E3 ubiquitin ligase, containing the BTB adaptor KLHL22, which regulates chromosome alignment and PLK1 kinetochore localization but not PLK1 stability. In the absence of KLHL22, PLK1 accumulates on kinetochores, resulting in activation of the spindle assembly checkpoint (SAC). CUL3-KLHL22 ubiquitylates Lys 492, located within the PBD, leading to PLK1 dissociation from kinetochore phosphoreceptors. Expression of a non-ubiquitylatable PLK1-K492R mutant phenocopies inactivation of CUL3-KLHL22. KLHL22 associates with the mitotic spindle and its interaction with PLK1 increases on chromosome bi-orientation. Our data suggest that CUL3-KLHL22-mediated ubiquitylation signals degradation-independent removal of PLK1 from kinetochores and SAC satisfaction, which are required for faithful mitosis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Amino Acid Sequence
- Blotting, Western
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Centrosome/metabolism
- Chromosomes, Human/genetics
- Cullin Proteins/antagonists & inhibitors
- Cullin Proteins/genetics
- Cullin Proteins/metabolism
- HeLa Cells
- Humans
- Immunoprecipitation
- Kinetochores/metabolism
- Microscopy, Fluorescence
- Microtubules/metabolism
- Mitosis/physiology
- Molecular Sequence Data
- Phosphorylation
- Protein Array Analysis
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Sequence Homology, Amino Acid
- Signal Transduction
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Spindle Apparatus/metabolism
- Ubiquitin/metabolism
- Ubiquitination
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Jochen Beck
- Institute of Biochemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Sarah Maerki
- Institute of Biochemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Markus Posch
- Wellcome Trust Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - Thibaud Metzger
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
| | | | | | - Kay Hofmann
- Institute for Genetics, University of Cologne, Cologne, Germany
| | | | | | - Jason R. Swedlow
- Wellcome Trust Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee, Scotland
| | - Matthias Peter
- Institute of Biochemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Izabela Sumara
- Institute of Biochemistry, ETH Zurich, 8093 Zurich, Switzerland
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
| |
Collapse
|
24
|
Bassermann F, Eichner R, Pagano M. The ubiquitin proteasome system - implications for cell cycle control and the targeted treatment of cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:150-62. [PMID: 23466868 DOI: 10.1016/j.bbamcr.2013.02.028] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/07/2013] [Accepted: 02/22/2013] [Indexed: 01/21/2023]
Abstract
Two families of E3 ubiquitin ligases are prominent in cell cycle regulation and mediate the timely and precise ubiquitin-proteasome-dependent degradation of key cell cycle proteins: the SCF (Skp1/Cul1/F-box protein) complex and the APC/C (anaphase promoting complex or cyclosome). While certain SCF ligases drive cell cycle progression throughout the cell cycle, APC/C (in complex with either of two substrate recruiting proteins: Cdc20 and Cdh1) orchestrates exit from mitosis (APC/C(Cdc20)) and establishes a stable G1 phase (APC/C(Cdh1)). Upon DNA damage or perturbation of the normal cell cycle, both ligases are involved in checkpoint activation. Mechanistic insight into these processes has significantly improved over the last ten years, largely due to a better understanding of APC/C and the functional characterization of multiple F-box proteins, the variable substrate recruiting components of SCF ligases. Here, we review the role of SCF- and APC/C-mediated ubiquitylation in the normal and perturbed cell cycle and discuss potential clinical implications of SCF and APC/C functions. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
Affiliation(s)
- Florian Bassermann
- Department of Medicine III, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany.
| | | | | |
Collapse
|
25
|
Campello L, Esteve-Rudd J, Cuenca N, Martín-Nieto J. The ubiquitin-proteasome system in retinal health and disease. Mol Neurobiol 2013; 47:790-810. [PMID: 23339020 DOI: 10.1007/s12035-012-8391-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
Abstract
The ubiquitin-proteasome system (UPS) is the main intracellular pathway for modulated protein turnover, playing an important role in the maintenance of cellular homeostasis. It also exerts a protein quality control through degradation of oxidized, mutant, denatured, or misfolded proteins and is involved in many biological processes where protein level regulation is necessary. This system allows the cell to modulate its protein expression pattern in response to changing physiological conditions and provides a critical protective role in health and disease. Impairments of UPS function in the central nervous system (CNS) underlie an increasing number of genetic and idiopathic diseases, many of which affect the retina. Current knowledge on the UPS composition and function in this tissue, however, is scarce and dispersed. This review focuses on UPS elements reported in the retina, including ubiquitinating and deubiquitinating enzymes (DUBs), and alternative proteasome assemblies. Known and inferred roles of protein ubiquitination, and of the related, SUMO conjugation (SUMOylation) process, in normal retinal development and adult homeostasis are addressed, including modulation of the visual cycle and response to retinal stress and injury. Additionally, the relationship between UPS dysfunction and human neurodegenerative disorders affecting the retina, including Alzheimer's, Parkinson's, and Huntington's diseases, are dealt with, together with numerous instances of retina-specific illnesses with UPS involvement, such as retinitis pigmentosa, macular degenerations, glaucoma, diabetic retinopathy (DR), and aging-related impairments. This information, though still basic and limited, constitutes a suitable framework to be expanded in incoming years and should prove orientative toward future therapy design targeting sight-affecting diseases with a UPS underlying basis.
Collapse
Affiliation(s)
- Laura Campello
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante, 03080 Alicante, Spain
| | | | | | | |
Collapse
|
26
|
Juranić M, Srilunchang KO, Krohn NG, Leljak-Levanić D, Sprunck S, Dresselhaus T. Germline-specific MATH-BTB substrate adaptor MAB1 regulates spindle length and nuclei identity in maize. THE PLANT CELL 2012; 24:4974-91. [PMID: 23250449 PMCID: PMC3556970 DOI: 10.1105/tpc.112.107169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 11/05/2012] [Accepted: 11/28/2012] [Indexed: 05/03/2023]
Abstract
Germline and early embryo development constitute ideal model systems to study the establishment of polarity, cell identity, and asymmetric cell divisions (ACDs) in plants. We describe here the function of the MATH-BTB domain protein MAB1 that is exclusively expressed in the germ lineages and the zygote of maize (Zea mays). mab1 (RNA interference [RNAi]) mutant plants display chromosome segregation defects and short spindles during meiosis that cause insufficient separation and migration of nuclei. After the meiosis-to-mitosis transition, two attached nuclei of similar identity are formed in mab1 (RNAi) mutants leading to an arrest of further germline development. Transient expression studies of MAB1 in tobacco (Nicotiana tabacum) Bright Yellow-2 cells revealed a cell cycle-dependent nuclear localization pattern but no direct colocalization with the spindle apparatus. MAB1 is able to form homodimers and interacts with the E3 ubiquitin ligase component Cullin 3a (CUL3a) in the cytoplasm, likely as a substrate-specific adapter protein. The microtubule-severing subunit p60 of katanin was identified as a candidate substrate for MAB1, suggesting that MAB1 resembles the animal key ACD regulator Maternal Effect Lethal 26 (MEL-26). In summary, our findings provide further evidence for the importance of posttranslational regulation for asymmetric divisions and germline progression in plants and identified an unstable key protein that seems to be involved in regulating the stability of a spindle apparatus regulator(s).
Collapse
Affiliation(s)
- Martina Juranić
- Cell Biology and Plant Biochemistry, Biochemie-Zentrum Regensburg, University of Regensburg, 93053 Regensburg, Germany
- Department of Molecular Biology, Faculty of Science and Mathematics, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Nádia Graciele Krohn
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirao Preto 14040-903, Brazil
| | - Dunja Leljak-Levanić
- Department of Molecular Biology, Faculty of Science and Mathematics, University of Zagreb, 10000 Zagreb, Croatia
| | - Stefanie Sprunck
- Cell Biology and Plant Biochemistry, Biochemie-Zentrum Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Thomas Dresselhaus
- Cell Biology and Plant Biochemistry, Biochemie-Zentrum Regensburg, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
27
|
Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat Cell Biol 2012; 14:117-23. [PMID: 22298039 DOI: 10.1038/ncb2407] [Citation(s) in RCA: 657] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ATP-driven chaperone valosin-containing protein (VCP)/p97 governs critical steps in ubiquitin-dependent protein quality control and intracellular signalling pathways. It cooperates with diverse partner proteins to help process ubiquitin-labelled proteins for recycling or degradation by the proteasome in many cellular contexts. Recent studies have uncovered unexpected cellular functions for p97 in autophagy, endosomal sorting and regulating protein degradation at the outer mitochondrial membrane, and elucidated a role for p97 in key chromatin-associated processes. These findings extend the functional relevance of p97 to lysosomal degradation and reveal a surprising dual role in protecting cells from protein stress and ensuring genome stability during proliferation.
Collapse
|
28
|
von Klitzing C, Huss R, Illert AL, Fröschl A, Wötzel S, Peschel C, Bassermann F, Duyster J. APC/C(Cdh1)-mediated degradation of the F-box protein NIPA is regulated by its association with Skp1. PLoS One 2011; 6:e28998. [PMID: 22205987 PMCID: PMC3243670 DOI: 10.1371/journal.pone.0028998] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 11/19/2011] [Indexed: 01/06/2023] Open
Abstract
NIPA (Nuclear Interaction Partner of Alk kinase) is an F-box like protein
that targets nuclear Cyclin B1 for degradation. Integrity and therefore activity
of the SCFNIPA E3 ligase is regulated by cell-cycle-dependent phosphorylation
of NIPA, restricting substrate ubiquitination to interphase. Here we show
that phosphorylated NIPA is degraded in late mitosis in an APC/CCdh1-dependent
manner. Binding of the unphosphorylated form of NIPA to Skp1 interferes with
binding to the APC/C-adaptor protein Cdh1 and therefore protects unphosphorylated
NIPA from degradation in interphase. Our data thus define a novel mode of
regulating APC/C-mediated ubiquitination.
Collapse
Affiliation(s)
| | - Richard Huss
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
| | - Anna Lena Illert
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
| | - Astrid Fröschl
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
| | - Sabine Wötzel
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
| | - Christian Peschel
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
| | - Justus Duyster
- Department of Internal Medicine ΙΙΙ,
Technical University of Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
29
|
Arjumand W, Sultana S. Role of VHL gene mutation in human renal cell carcinoma. Tumour Biol 2011; 33:9-16. [PMID: 22125026 DOI: 10.1007/s13277-011-0257-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 10/25/2011] [Indexed: 12/17/2022] Open
Abstract
The Von Hippel-Lindau (VHL) is an inherited neoplasia syndrome caused by the inactivation of VHL tumor suppressor gene, and somatic mutation of this gene has been related to the development of sporadic clear cell renal carcinoma. The affected individuals are at higher risk for the development of tumor in other organs, which include pheochromocytomas, retinal angioma, pancreatic cysts, and CNS hemangioblastomas. The VHL mRNA encodes a protein (pVHL) that contains 213 amino acid residues which migrate with an apparent molecular weight of 24 to 30 kDa. The VHL gene protein has multiple functions that are linked to tumor suppression, but the best recognized and evidently linked to the development of renal cell carcinoma (RCC) is inhibition of hypoxia-inducible factor (HIF), as well as plays a role in targeting HIF for ubiquitin-mediated degradation. Aberrations in VHL's function, either through mutation or promoter hypermethylation, lead to the accumulation of HIF, which will transcriptionally upregulate a sequence of hypoxia responsive genes, including epidermal growth factor, vascular endothelial growth factor, platelet-derived growth factor, and other proangiogenic factors, resulting in upregulated blood vessel growth, one of the prerequisites of a tumor. HIF plays a critical role in pVHL-defective tumor formation, raising the possibility that drugs directed against HIF or its downstream targets (such as vascular endothelial growth factor) may one day play a role in the treatment of RCC. Moreover, a number of drugs have been developed that target HIF-responsive gene products, many of these targeted therapies have demonstrated significant activity in kidney cancer clinical trials and signify substantive advances in the treatment of this disease.
Collapse
Affiliation(s)
- Wani Arjumand
- Section of Molecular Carcinogenesis and Chemoprevention, Department of Medical Elementology and Toxicology, Faculty of Science, Jamia Hamdard, Hamdard University, Hamdard Nagar, New Delhi 110062, India.
| | | |
Collapse
|
30
|
Dobrynin G, Popp O, Romer T, Bremer S, Schmitz MHA, Gerlich DW, Meyer H. Cdc48/p97-Ufd1-Npl4 antagonizes Aurora B during chromosome segregation in HeLa cells. J Cell Sci 2011; 124:1571-80. [PMID: 21486945 DOI: 10.1242/jcs.069500] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
During exit from mitosis in Xenopus laevis egg extracts, the AAA+ ATPase Cdc48/p97 (also known as VCP in vertebrates) and its adapter Ufd1-Npl4 remove the kinase Aurora B from chromatin to allow nucleus formation. Here, we show that in HeLa cells Ufd1-Npl4 already antagonizes Aurora B on chromosomes during earlier mitotic stages and that this is crucial for proper chromosome segregation. Depletion of Ufd1-Npl4 by small interfering RNA (siRNA) caused chromosome alignment and anaphase defects resulting in missegregated chromosomes and multi-lobed nuclei. Ufd1-Npl4 depletion also led to increased levels of Aurora B on prometaphase and metaphase chromosomes. This increase was associated with higher Aurora B activity, as evidenced by the partial resistance of CENP-A phosphorylation to the Aurora B inhibitor hesperadin. Furthermore, low concentrations of hesperadin partially rescued chromosome alignment in Ufd1-depleted cells, whereas, conversely, Ufd1-depletion partially restored congression in the presence of hesperadin. These data establish Cdc48/p97-Ufd1-Npl4 as a crucial negative regulator of Aurora B early in mitosis of human somatic cells and suggest that the activity of Aurora B on chromosomes needs to be restrained to ensure faithful chromosome segregation.
Collapse
Affiliation(s)
- Grzegorz Dobrynin
- Centre for Medical Biotechnology, University of Duisburg-Essen, 45117 Essen, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
Cui Y, Cheng X, Zhang C, Zhang Y, Li S, Wang C, Guadagno TM. Degradation of the human mitotic checkpoint kinase Mps1 is cell cycle-regulated by APC-cCdc20 and APC-cCdh1 ubiquitin ligases. J Biol Chem 2010; 285:32988-32998. [PMID: 20729194 DOI: 10.1074/jbc.m110.140905] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mps1 is a dual specificity protein kinase with key roles in regulating the spindle assembly checkpoint and chromosome-microtubule attachments. Consistent with these mitotic functions, Mps1 protein levels fluctuate during the cell cycle, peaking at early mitosis and abruptly declining during mitotic exit and progression into the G(1) phase. Although evidence in budding yeast indicates that Mps1 is targeted for degradation at anaphase by the anaphase-promoting complex (APC)-c(Cdc20) complex, little is known about the regulatory mechanisms that govern Mps1 protein levels in human cells. Here, we provide evidence for the ubiquitin ligase/proteosome pathway in regulating human Mps1 levels during late mitosis through G(1) phase. First, we showed that treatment of HEK 293T cells with the proteosome inhibitor MG132 resulted in an increase in both the polyubiquitination and the accumulation of Mps1 protein levels. Next, Mps1 was shown to co-precipitate with APC and its activators Cdc20 and Cdh1 in a cell cycle-dependent manner. Consistent with this, overexpression of Cdc20 or Cdh1 led to a marked reduction of endogenous Mps1 levels during anaphase or G(1) phase, respectively. In contrast, depletion of Cdc20 or Cdh1 by RNAi treatment both led to the stabilization of Mps1 protein during mitosis or G(1) phase, respectively. Finally, we identified a single D-box motif in human Mps1 that is required for its ubiquitination and degradation. Failure to appropriately degrade Mps1 is sufficient to trigger centrosome amplification and mitotic abnormalities in human cells. Thus, our results suggest that the sequential actions of the APC-c(Cdc20) and APC-c(Cdh1) ubiquitin ligases regulate the clearance of Mps1 levels and are critical for Mps1 functions during the cell cycle in human cells.
Collapse
Affiliation(s)
- Yongping Cui
- From the Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan, Shanxi 030001, China; Department of Cell Biology and Genetics, Taiyuan, Shanxi 030001, China.
| | - Xiaolong Cheng
- Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ce Zhang
- From the Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan, Shanxi 030001, China
| | - Yanyan Zhang
- Department of Cell Biology and Genetics, Taiyuan, Shanxi 030001, China
| | - Shujing Li
- Department of Cell Biology and Genetics, Taiyuan, Shanxi 030001, China
| | - Chuangui Wang
- School of Life Science, East China Normal University, Shanghai 200062, China
| | - Thomas M Guadagno
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612.
| |
Collapse
|
32
|
Liu M, Schmidt EE, Halford WP. ICP0 dismantles microtubule networks in herpes simplex virus-infected cells. PLoS One 2010; 5:e10975. [PMID: 20544015 PMCID: PMC2882321 DOI: 10.1371/journal.pone.0010975] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 05/13/2010] [Indexed: 01/18/2023] Open
Abstract
Infected-cell protein 0 (ICP0) is a RING finger E3 ligase that regulates herpes simplex virus (HSV) mRNA synthesis, and strongly influences the balance between latency and replication of HSV. For 25 years, the nuclear functions of ICP0 have been the subject of intense scrutiny. To obtain new clues about ICP0's mechanism of action, we constructed HSV-1 viruses that expressed GFP-tagged ICP0. To our surprise, both GFP-tagged and wild-type ICP0 were predominantly observed in the cytoplasm of HSV-infected cells. Although ICP0 is exclusively nuclear during the immediate-early phase of HSV infection, further analysis revealed that ICP0 translocated to the cytoplasm during the early phase where it triggered a previously unrecognized process; ICP0 dismantled the microtubule network of the host cell. A RING finger mutant of ICP0 efficiently bundled microtubules, but failed to disperse microtubule bundles. Synthesis of ICP0 proved to be necessary and sufficient to disrupt microtubule networks in HSV-infected and transfected cells. Plant and animal viruses encode many proteins that reorganize microtubules. However, this is the first report of a viral E3 ligase that regulates microtubule stability. Intriguingly, several cellular E3 ligases orchestrate microtubule disassembly and reassembly during mitosis. Our results suggest that ICP0 serves a dual role in the HSV life cycle, acting first as a nuclear regulator of viral mRNA synthesis and acting later, in the cytoplasm, to dismantle the host cell's microtubule network in preparation for virion synthesis and/or egress.
Collapse
Affiliation(s)
- Mingyu Liu
- Department of Microbiology and Immunology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Edward E. Schmidt
- Department of Veterinary Molecular Biology, Montana State University, Bozeman, Montana, United States of America
| | - William P. Halford
- Department of Microbiology and Immunology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| |
Collapse
|
33
|
Pawar SA, Sarkar TR, Balamurugan K, Sharan S, Wang J, Zhang Y, Dowdy SF, Huang AM, Sterneck E. C/EBP{delta} targets cyclin D1 for proteasome-mediated degradation via induction of CDC27/APC3 expression. Proc Natl Acad Sci U S A 2010; 107:9210-5. [PMID: 20439707 PMCID: PMC2889124 DOI: 10.1073/pnas.0913813107] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The transcription factor CCAAT/enhancer binding protein delta (C/EBPdelta, CEBPD, NFIL-6beta) has tumor suppressor function; however, the molecular mechanism(s) by which C/EBPdelta exerts its effect are largely unknown. Here, we report that C/EBPdelta induces expression of the Cdc27 (APC3) subunit of the anaphase promoting complex/cyclosome (APC/C), which results in the polyubiquitination and degradation of the prooncogenic cell cycle regulator cyclin D1, and also down-regulates cyclin B1, Skp2, and Plk-1. In C/EBPdelta knockout mouse embryo fibroblasts (MEF) Cdc27 levels were reduced, whereas cyclin D1 levels were increased even in the presence of activated GSK-3beta. Silencing of C/EBPdelta, Cdc27, or the APC/C coactivator Cdh1 (FZR1) in MCF-10A breast epithelial cells increased cyclin D1 protein expression. Like C/EBPdelta, and in contrast to cyclin D1, Cdc27 was down-regulated in several breast cancer cell lines, suggesting that Cdc27 itself may be a tumor suppressor. Cyclin D1 is a known substrate of polyubiquitination complex SKP1/CUL1/F-box (SCF), and our studies show that Cdc27 directs cyclin D1 to alternative degradation by APC/C. These findings shed light on the role and regulation of APC/C, which is critical for most cellular processes.
Collapse
Affiliation(s)
- Snehalata A. Pawar
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Tapasree Roy Sarkar
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Kuppusamy Balamurugan
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Shikha Sharan
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Jun Wang
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Youhong Zhang
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Steven F. Dowdy
- Department of Cellular and Molecular Medicine, Howard Hughes Medical Institute, University of California, San Diego School of Medicine, La Jolla, CA 92093-0686
| | - A-Mei Huang
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| | - Esta Sterneck
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201; and
| |
Collapse
|
34
|
Xing H, Hong Y, Sarge KD. PEST sequences mediate heat shock factor 2 turnover by interacting with the Cul3 subunit of the Cul3-RING ubiquitin ligase. Cell Stress Chaperones 2010; 15:301-8. [PMID: 19768582 PMCID: PMC2866995 DOI: 10.1007/s12192-009-0144-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 08/31/2009] [Indexed: 11/27/2022] Open
Abstract
Cullin-RING ubiquitin ligases promote the polyubiquitination and degradation of many important cellular proteins, which previous studies indicated can be targeted for degradation via interaction with BTB domain-containing subunits of this E3 ligase complex. PEST domains are known to promote the degradation of proteins that contain them. However, the molecular mechanism by which PEST sequences promote degradation of these proteins is not understood. Here we show that the PEST sequences of a short-lived protein called HSF2 interact with Cullin3, a subunit of a Cullin-RING E3 ubiquitin ligase, and that this interaction mediates the Cul3-dependent ubiquitination and degradation of HSF2. These results indicate how, at the molecular level, PEST sequences can promote the proteolysis of proteins that contain them. They also expand understanding of the mechanisms by which substrates can be recruited to Cullin-RING E3 ubiquitin ligases to include interactions between PEST sequences and Cul3.
Collapse
Affiliation(s)
- Hongyan Xing
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S. Limestone Street, Lexington, KY 40536 USA
| | - Yiling Hong
- Department of Biology, University of Dayton, Dayton, OH 45469 USA
| | - Kevin D. Sarge
- Department of Molecular and Cellular Biochemistry, University of Kentucky, 741 S. Limestone Street, Lexington, KY 40536 USA
| |
Collapse
|
35
|
Abstract
Recent studies have demonstrated that a number of E3 ubiquitin ligases, including Cbl, Smurf1, Smurf2, HDM2, BCA2, SCF(beta-TRCP) and XRNF185, play important roles in cell adhesion and migration. Cbl negatively regulates cell adhesion via alpha integrin and Rap1 and inhibits actin polymerization by ubiquitinating mDab1 and WAVE2. Smurf1 regulates cell migration through ubiquitination of RhoA, talin head domain and hPEM2, while Smurf2 ubiquitinates Smurf1, TGFbeta type I receptor and RaplB to modulate cell migration and adhesion. HDM2 negatively regulates cell migration by targeting NFAT (a transcription factor) for ubiquitination and degradation, while SCF(beta-TRCP) ubiquitinates Snail (a transcriptional repressor of E-cadherin) to inhibit cell migration. TRIM32 promotes cell migration through ubiquitination of Abl interactor 2 (Abi2), a tumor suppressor. RNF5 and XRNF185 modulate cell migration by ubiquitinating paxillin. Thus, these E3 ubiquitin ligases regulate cell adhesion and (or) migration through ubiquitination of their specific substrates.
Collapse
Affiliation(s)
- Cai Huang
- Department of Cell and Developmental Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
36
|
Maerki S, Olma MH, Staubli T, Steigemann P, Gerlich DW, Quadroni M, Sumara I, Peter M. The Cul3-KLHL21 E3 ubiquitin ligase targets aurora B to midzone microtubules in anaphase and is required for cytokinesis. ACTA ACUST UNITED AC 2010; 187:791-800. [PMID: 19995937 PMCID: PMC2806313 DOI: 10.1083/jcb.200906117] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Selective ubiquitination of Aurora B by different Cul3 adaptors targets it at the correct time to the correct place during mitosis. Cul3 (Cullin3)-based E3 ubiquitin ligases recently emerged as critical regulators of mitosis. In this study, we identify two mammalian BTB (Bric-a-brac–Tramtrack–Broad complex)-Kelch proteins, KLHL21 and KLHL22, that interact with Cul3 and are required for efficient chromosome alignment. Interestingly, KLHL21 but not KLHL22 is necessary for cytokinesis and regulates translocation of the chromosomal passenger complex (CPC) from chromosomes to the spindle midzone in anaphase, similar to the previously described BTB-Kelch proteins KLHL9 and KLHL13. KLHL21 directly binds to Aurora B and mediates ubiquitination of Aurora B in vitro. In contrast to KLHL9 and KLHL13, KLHL21 localizes to midzone microtubules in anaphase and recruits Aurora B and Cul3 to this region. Together, our results suggest that different Cul3 adaptors nonredundantly regulate Aurora B during mitosis, possibly by ubiquitinating different pools of Aurora B at distinct subcellular localizations.
Collapse
Affiliation(s)
- Sarah Maerki
- Institute of Biochemistry, Swiss Federal Institute of Technology Zurich, CH-8093 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Cullin neddylation and substrate-adaptors counteract SCF inhibition by the CAND1-like protein Lag2 in Saccharomyces cerevisiae. EMBO J 2010; 28:3845-56. [PMID: 19942853 DOI: 10.1038/emboj.2009.354] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 11/05/2009] [Indexed: 11/08/2022] Open
Abstract
Cullin-based E3 ubiquitin ligases are activated through covalent modification of the cullin subunit by the ubiquitin-like protein Nedd8. Cullin neddylation dissociates the ligase assembly inhibitor Cand1, and promotes E2 recruitment and ubiquitin transfer by inducing a conformational change. Here, we have identified and characterized Lag2 as a likely Saccharomyces cerevisiae orthologue of mammalian Cand1. Similar to Cand1, Lag2 directly interacts with non-neddylated yeast cullin Cdc53 and prevents its neddylation in vivo and in vitro. Binding occurs through a conserved C-terminal beta-hairpin structure that inserts into the Skp1-binding pocket on the cullin, and an N-terminal motif that covers the neddylation lysine. Interestingly, Lag2 is itself neddylated in vivo on a lysine adjacent to this N-terminal-binding site. Overexpression of Lag2 inhibits Cdc53 activity in strains defective for Skp1 or neddylation functions, implying that these activities are important to counteract Lag2 in vivo. Our results favour a model in which binding of substrate-specific adaptors triggers release of Cand1/Lag2, whereas subsequent neddylation of the cullin facilitates the removal and prevents re-association of Lag2/Cand1.
Collapse
|
38
|
Abstract
Ubiquitylation of caspase-8 by the Cullin3 E3 ligase allows its translocation to cytosolic aggregates in the cell by p62/sequestosome-1, increasing caspase-8 activation and thus leading to TRAIL-induced cell death.
Collapse
Affiliation(s)
- W Wei-Lynn Wong
- Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, VIC 3086, Australia.
| | | |
Collapse
|
39
|
Ban R, Matsuzaki H, Akashi T, Sakashita G, Taniguchi H, Park SY, Tanaka H, Furukawa K, Urano T. Mitotic regulation of the stability of microtubule plus-end tracking protein EB3 by ubiquitin ligase SIAH-1 and Aurora mitotic kinases. J Biol Chem 2009; 284:28367-28381. [PMID: 19696028 DOI: 10.1074/jbc.m109.000273] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Microtubule plus-end tracking proteins (+TIPs) control microtubule dynamics in fundamental processes such as cell cycle, intracellular transport, and cell motility, but how +TIPs are regulated during mitosis remains largely unclear. Here we show that the endogenous end-binding protein family EB3 is stable during mitosis, facilitates cell cycle progression at prometaphase, and then is down-regulated during the transition to G(1) phase. The ubiquitin-protein isopeptide ligase SIAH-1 facilitates EB3 polyubiquitination and subsequent proteasome-mediated degradation, whereas SIAH-1 knockdown increases EB3 stability and steady-state levels. Two mitotic kinases, Aurora-A and Aurora-B, phosphorylate endogenous EB3 at Ser-176, and the phosphorylation triggers disruption of the EB3-SIAH-1 complex, resulting in EB3 stabilization during mitosis. Our results provide new insight into a regulatory mechanism of +TIPs in cell cycle transition.
Collapse
Affiliation(s)
- Reiko Ban
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501; Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550.
| | - Hideki Matsuzaki
- Institute for Enzyme Research, University of Tokushima, 3-15-18 Kuramoto, Tokushima 770-8503
| | - Tomohiro Akashi
- Division of Molecular Mycology and Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550
| | - Gyosuke Sakashita
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501; Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550
| | - Hisaaki Taniguchi
- Institute for Enzyme Research, University of Tokushima, 3-15-18 Kuramoto, Tokushima 770-8503
| | - Sam-Yong Park
- Protein Design Laboratory, Yokohama City University, Tsurumi, Yokohama 230-0045
| | - Hirofumi Tanaka
- School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo 192-0392, Japan
| | - Koichi Furukawa
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550
| | - Takeshi Urano
- Department of Biochemistry, Shimane University School of Medicine, Izumo 693-8501; Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550.
| |
Collapse
|
40
|
Friedman JS, Ray JW, Waseem N, Johnson K, Brooks MJ, Hugosson T, Breuer D, Branham KE, Krauth DS, Bowne SJ, Sullivan LS, Ponjavic V, Gränse L, Khanna R, Trager EH, Gieser LM, Hughbanks-Wheaton D, Cojocaru RI, Ghiasvand NM, Chakarova CF, Abrahamson M, Göring HH, Webster AR, Birch DG, Abecasis GR, Fann Y, Bhattacharya SS, Daiger SP, Heckenlively JR, Andréasson S, Swaroop A. Mutations in a BTB-Kelch protein, KLHL7, cause autosomal-dominant retinitis pigmentosa. Am J Hum Genet 2009; 84:792-800. [PMID: 19520207 DOI: 10.1016/j.ajhg.2009.05.007] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 05/19/2009] [Accepted: 05/20/2009] [Indexed: 01/08/2023] Open
Abstract
Retinitis pigmentosa (RP) refers to a genetically heterogeneous group of progressive neurodegenerative diseases that result in dysfunction and/or death of rod and cone photoreceptors in the retina. So far, 18 genes have been identified for autosomal-dominant (ad) RP. Here, we describe an adRP locus (RP42) at chromosome 7p15 through linkage analysis in a six-generation Scandinavian family and identify a disease-causing mutation, c.449G-->A (p.S150N), in exon 6 of the KLHL7 gene. Mutation screening of KLHL7 in 502 retinopathy probands has revealed three different missense mutations in six independent families. KLHL7 is widely expressed, including expression in rod photoreceptors, and encodes a 75 kDa protein of the BTB-Kelch subfamily within the BTB superfamily. BTB-Kelch proteins have been implicated in ubiquitination through Cullin E3 ligases. Notably, all three putative disease-causing KLHL7 mutations are within a conserved BACK domain; homology modeling suggests that mutant amino acid side chains can potentially fill the cleft between two helices, thereby affecting the ubiquitination complexes. Mutations in an identical region of another BTB-Kelch protein, gigaxonin, have previously been associated with giant axonal neuropathy. Our studies suggest an additional role of the ubiquitin-proteasome protein-degradation pathway in maintaining neuronal health and in disease.
Collapse
|
41
|
Think locally: control of ubiquitin-dependent protein degradation in neurons. EMBO Rep 2008; 10:44-50. [PMID: 19079132 DOI: 10.1038/embor.2008.229] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 11/13/2008] [Indexed: 11/08/2022] Open
Abstract
The nervous system coordinates many aspects of body function such as learning, memory, behaviour and locomotion. Therefore, it must develop and maintain an intricate network of differentiated neuronal cells, which communicate efficiently with each other and with non-neuronal target cells. Unlike most somatic cells, differentiated neurons are post-mitotic and characterized by a highly polarized morphology that determines the flow of information. Among other post-translational modifications, the ubiquitination of specific protein substrates was recently shown to have a crucial role in the regulation of neuronal development and differentiation. Here, we review recent findings that illustrate the mechanisms that mediate the temporal and spatial control of neuronal protein turnover by the ubiquitin-proteasome system (UPS), which is crucial for the development and function of the nervous system.
Collapse
|