1
|
Lee HC, Hsieh CC, Tsai HJ. KEPI plays a negative role in the repression that accompanies translational inhibition guided by the uORF element of human CHOP transcript during stress response. Gene X 2022; 817:146160. [PMID: 35031423 DOI: 10.1016/j.gene.2021.146160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/28/2021] [Accepted: 12/10/2021] [Indexed: 11/04/2022] Open
Abstract
Translation of the downstream coding sequence of some mRNAs may be repressed by the upstream open reading frame (uORF) at their 5'-end. The mechanism underlying this uORF-mediated translational inhibition (uORF-MTI) is not fully understood in vivo. Recently, it was found that zebrafish Endouc or its human orthologue ENDOU (Endouc/ENDOU) plays a positive role in repressing the uORF-MTI of human CHOP (uORFchop-MTI) during stress by blocking its activity However, the repression of uORFchop-MTI assisted by an as-yet unidentified negative effector remains to be elucidated. Compared to the upregulated CHOP transcript, we herein report that the kepi (kinase-enhanced PP1 inhibitor) transcript was downregulated in the zebrafish embryos treated with both heat shock and hypoxia. Quantitative RT-PCR also revealed that the level of kepi mRNA was noticeably decreased in both heat-shock-treated and hypoxia-exposed embryos. When kepi mRNA was microinjected into the one-celled embryos from transgenic line huORFZ, the translation of downstream GFP reporter controlled by the uORFchop-MTI was reduced in the hypoxia-exposed embryos. In contrast, when kepi was knocked down by injection of antisense Morpholino oligonucleotide, the translation of downstream GFP reporter was induced and expressed in the brain and spinal cord of injected embryos in the absence of stress. During normal condition, overexpression of KEPI increased eIF2α phosphorylation, resulting in inducing the translation of uORF-tag mRNA, such as ATF4 and CHOP mRNAs. However, during stress condition, overexpression of KEPI decreased eIF2α phosphorylation, resulting in reducing the GFP reporter and CHOP proteins. This is the first report to demonstrate that KEPI plays a negative role in uORFchop - mediated translation during ER stress.
Collapse
Affiliation(s)
- Hung-Chieh Lee
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Chi-Cheng Hsieh
- The Liver Disease Prevention and Treatment Research Foundation, Taipei, Taiwan
| | - Huai-Jen Tsai
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan; School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
2
|
Jian Y, Kong L, Xu H, Shi Y, Huang X, Zhong W, Huang S, Li Y, Shi D, Xiao Y, Yang M, Li S, Chen X, Ouyang Y, Hu Y, Chen X, Song L, Ye R, Wei W. Protein phosphatase 1 regulatory inhibitor subunit 14C promotes triple-negative breast cancer progression via sustaining inactive glycogen synthase kinase 3 beta. Clin Transl Med 2022; 12:e725. [PMID: 35090098 PMCID: PMC8797469 DOI: 10.1002/ctm2.725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is fast-growing and highly metastatic with the poorest prognosis among the breast cancer subtypes. Inactivation of glycogen synthase kinase 3 beta (GSK3β) plays a vital role in the aggressiveness of TNBC; however, the underlying mechanism for sustained GSK3β inhibition remains largely unknown. Here, we find that protein phosphatase 1 regulatory inhibitor subunit 14C (PPP1R14C) is upregulated in TNBC and relevant to poor prognosis in patients. Overexpression of PPP1R14C facilitates cell proliferation and the aggressive phenotype of TNBC cells, whereas the depletion of PPP1R14C elicits opposite effects. Moreover, PPP1R14C is phosphorylated and activated by protein kinase C iota (PRKCI) at Thr73. p-PPP1R14C then represses Ser/Thr protein phosphatase type 1 (PP1) to retain GSK3β phosphorylation at high levels. Furthermore, p-PPP1R14C recruits E3 ligase, TRIM25, toward the ubiquitylation and degradation of non-phosphorylated GSK3β. Importantly, the blockade of PPP1R14C phosphorylation inhibits xenograft tumorigenesis and lung metastasis of TNBC cells. These findings provide a novel mechanism for sustained GSK3β inactivation in TNBC and suggest that PPP1R14C might be a potential therapeutic target.
Collapse
Affiliation(s)
- Yunting Jian
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Pathology, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Lingzhi Kong
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Hongyi Xu
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yawei Shi
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xinjian Huang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Wenjing Zhong
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yue Li
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Dongni Shi
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yunyun Xiao
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Muwen Yang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Siqi Li
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xiangfu Chen
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Ying Ouyang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yameng Hu
- Department of Biochemistry, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Xin Chen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences; Guangzhou Institute of OncologyTumor Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Libing Song
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Runyi Ye
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Weidong Wei
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
3
|
Sun S, Zane A, Fulton C, Philipoom J. Statistical and bioinformatic analysis of hemimethylation patterns in non-small cell lung cancer. BMC Cancer 2021; 21:268. [PMID: 33711952 PMCID: PMC7953768 DOI: 10.1186/s12885-021-07990-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/01/2021] [Indexed: 12/22/2022] Open
Abstract
Background DNA methylation is an epigenetic event involving the addition of a methyl-group to a cytosine-guanine base pair (i.e., CpG site). It is associated with different cancers. Our research focuses on studying non-small cell lung cancer hemimethylation, which refers to methylation occurring on only one of the two DNA strands. Many studies often assume that methylation occurs on both DNA strands at a CpG site. However, recent publications show the existence of hemimethylation and its significant impact. Therefore, it is important to identify cancer hemimethylation patterns. Methods In this paper, we use the Wilcoxon signed rank test to identify hemimethylated CpG sites based on publicly available non-small cell lung cancer methylation sequencing data. We then identify two types of hemimethylated CpG clusters, regular and polarity clusters, and genes with large numbers of hemimethylated sites. Highly hemimethylated genes are then studied for their biological interactions using available bioinformatics tools. Results In this paper, we have conducted the first-ever investigation of hemimethylation in lung cancer. Our results show that hemimethylation does exist in lung cells either as singletons or clusters. Most clusters contain only two or three CpG sites. Polarity clusters are much shorter than regular clusters and appear less frequently. The majority of clusters found in tumor samples have no overlap with clusters found in normal samples, and vice versa. Several genes that are known to be associated with cancer are hemimethylated differently between the cancerous and normal samples. Furthermore, highly hemimethylated genes exhibit many different interactions with other genes that may be associated with cancer. Hemimethylation has diverse patterns and frequencies that are comparable between normal and tumorous cells. Therefore, hemimethylation may be related to both normal and tumor cell development. Conclusions Our research has identified CpG clusters and genes that are hemimethylated in normal and lung tumor samples. Due to the potential impact of hemimethylation on gene expression and cell function, these clusters and genes may be important to advance our understanding of the development and progression of non-small cell lung cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07990-7.
Collapse
Affiliation(s)
- Shuying Sun
- Department of Mathematics, Texas State University, San Marcos, TX, USA.
| | - Austin Zane
- Department of Statistics, Texas A&M University, College Station, TX, USA
| | - Carolyn Fulton
- Department of Mathematics, Schreiner University, Kerrville, TX, USA
| | - Jasmine Philipoom
- Department of Mathematics, Applied Mathematics, and Statistics, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Casamayor A, Ariño J. Controlling Ser/Thr protein phosphatase PP1 activity and function through interaction with regulatory subunits. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 122:231-288. [PMID: 32951813 DOI: 10.1016/bs.apcsb.2020.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein phosphatase 1 is a major Ser/Thr protein phosphatase activity in eukaryotic cells. It is composed of a catalytic polypeptide (PP1C), with little substrate specificity, that interacts with a large variety of proteins of diverse structure (regulatory subunits). The diversity of holoenzymes that can be formed explain the multiplicity of cellular functions under the control of this phosphatase. In quite a few cases, regulatory subunits have an inhibitory role, downregulating the activity of the phosphatase. In this chapter we shall introduce PP1C and review the most relevant families of PP1C regulatory subunits, with particular emphasis in describing the structural basis for their interaction.
Collapse
Affiliation(s)
- Antonio Casamayor
- Institut de Biotecnologia i Biomedicina & Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola, del Vallès, Spain
| | - Joaquín Ariño
- Institut de Biotecnologia i Biomedicina & Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola, del Vallès, Spain
| |
Collapse
|
5
|
Dahal B, Lin SC, Carey BD, Jacobs JL, Dinman JD, van Hoek ML, Adams AA, Kehn-Hall K. EGR1 upregulation following Venezuelan equine encephalitis virus infection is regulated by ERK and PERK pathways contributing to cell death. Virology 2019; 539:121-128. [PMID: 31733451 PMCID: PMC7126400 DOI: 10.1016/j.virol.2019.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 10/02/2019] [Accepted: 10/28/2019] [Indexed: 01/05/2023]
Abstract
Venezuelan equine encephalitis virus (VEEV) is a neurotropic virus that causes significant disease in both humans and equines. Here we characterized the impact of VEEV on signaling pathways regulating cell death in human primary astrocytes. VEEV productively infected primary astrocytes and caused an upregulation of early growth response 1 (EGR1) gene expression at 9 and 18 h post infection. EGR1 induction was dependent on extracellular signal-regulated kinase1/2 (ERK1/2) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), but not on p38 mitogen activated protein kinase (MAPK) or phosphoinositide 3-kinase (PI3K) signaling. Knockdown of EGR1 significantly reduced VEEV-induced apoptosis and impacted viral replication. Knockdown of ERK1/2 or PERK significantly reduced EGR1 gene expression, dramatically reduced viral replication, and increased cell survival as well as rescued cells from VEEV-induced apoptosis. These data indicate that EGR1 activation and subsequent cell death are regulated through ERK and PERK pathways in VEEV infected primary astrocytes.
Collapse
Affiliation(s)
- Bibha Dahal
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Shih-Chao Lin
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Brian D Carey
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Jonathan L Jacobs
- QIAGEN Bioinformatics, Aarhus, Denmark; QIAGEN Bioinformatics, Maryland, USA
| | - Jonathan D Dinman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Monique L van Hoek
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | | | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
6
|
Xiong L, Xia WF, Tang FL, Pan JX, Mei L, Xiong WC. Retromer in Osteoblasts Interacts With Protein Phosphatase 1 Regulator Subunit 14C, Terminates Parathyroid Hormone's Signaling, and Promotes Its Catabolic Response. EBioMedicine 2016; 9:45-60. [PMID: 27333042 PMCID: PMC4972523 DOI: 10.1016/j.ebiom.2016.05.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/11/2016] [Accepted: 05/24/2016] [Indexed: 12/02/2022] Open
Abstract
Parathyroid hormone (PTH) plays critical, but distinct, roles in bone remodeling, including bone formation (anabolic response) and resorption (catabolic response). Although its signaling and function have been extensively investigated, it just began to be understood how distinct functions are induced by PTH activating a common receptor, the PTH type 1 receptor (PTH1R), and how PTH1R signaling is terminated. Here, we provide evidence for vacuolar protein sorting 35 (VPS35), a major component of retromer, in regulating PTH1R trafficking, turning off PTH signaling, and promoting its catabolic function. VPS35 is expressed in osteoblast (OB)-lineage cells. VPS35-deficiency in OBs impaired PTH(1–34)-promoted PTH1R translocation to the trans-Golgi network, enhanced PTH(1–34)-driven signaling, and reduced PTH(1–34)'s catabolic response in culture and in mice. Further mechanical studies revealed that VPS35 interacts with not only PTH1R, but also protein phosphatase 1 regulatory subunit 14C (PPP1R14C), an inhibitory subunit of PP1 phosphatase. PPP1R14C also interacts with PTH1R, which is necessary for the increased endosomal PTH1R signaling and decreased PTH(1–34)'s catabolic response in VPS35-deficient OB-lineage cells. Taken together, these results suggest that VPS35 deregulates PTH1R-signaling likely by its interaction with PTH1R and PPP1R14C. This event is critical for the control of PTH(1–34)-signaling dynamics, which may underlie PTH-induced catabolic response and adequate bone remodeling. VPS35 terminates PTH(1-34)-induced cell surface and endosomal signalings Osteoblastic VPS35 promotes PTH(1-34)-driven catabolic response VPS35 interacts with PPP1R14C PPP1R14C also interacts with PTH1R and promotes PTH(1-34)-induced endosomal signaling PPP1R14C is necessary for the increased endosomal PTH1R signaling and decreased PTH(1-34)’s catabolic response in VPS35-deficient OB-lineage cells
Collapse
Affiliation(s)
- Lei Xiong
- Department of Neuroscience & Regenerative Medicine, Department of Neurology, Medical College of Georgia, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Wen-Fang Xia
- Department of Neuroscience & Regenerative Medicine, Department of Neurology, Medical College of Georgia, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fu-Lei Tang
- Department of Neuroscience & Regenerative Medicine, Department of Neurology, Medical College of Georgia, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Jin-Xiu Pan
- Department of Neuroscience & Regenerative Medicine, Department of Neurology, Medical College of Georgia, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Lin Mei
- Department of Neuroscience & Regenerative Medicine, Department of Neurology, Medical College of Georgia, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - Wen-Cheng Xiong
- Department of Neuroscience & Regenerative Medicine, Department of Neurology, Medical College of Georgia, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States.
| |
Collapse
|
7
|
Dedinszki D, Kiss A, Márkász L, Márton A, Tóth E, Székely L, Erdődi F. Inhibition of protein phosphatase-1 and -2A decreases the chemosensitivity of leukemic cells to chemotherapeutic drugs. Cell Signal 2014; 27:363-72. [PMID: 25435424 DOI: 10.1016/j.cellsig.2014.11.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/21/2014] [Indexed: 01/27/2023]
Abstract
The phosphorylation of key proteins balanced by protein kinases and phosphatases are implicated in the regulation of cell cycle and apoptosis of malignant cells and influences anticancer drug actions. The efficacy of daunorubicin (DNR) in suppression of leukemic cell survival was investigated in the presence of tautomycin (TM) and calyculin A (CLA), specific membrane permeable inhibitors of protein phosphatase-1 (PP1) and -2A (PP2A), respectively. CLA (50 nM) or TM (1μM) suppressed viability of THP-1 and KG-1 myeloid leukemia cell lines to moderate extents; however, they significantly increased survival upon DNR-induced cell death. CLA increased the phosphorylation level of Erk1/2 and PKB/Akt kinases, the retinoblastoma protein (pRb), decreased caspase-3 activation by DNR and increased the phosphorylation level of the inhibitory sites (Thr696 and Thr853) in the myosin phosphatase (MP) target subunit (MYPT1) as well as in a 25kDa kinase-enhanced phosphatase inhibitor (KEPI)-like protein. TM induced enhanced phosphorylation of pRb only, suggesting that this event may be a common factor upon CLA-induced PP2A and TM-induced PP1 inhibitory influences on cell survival. Silencing PP1 by siRNA in HeLa cells, or overexpression of Flag-KEPI in MCF-7 cells coupled with inducing its phosphorylation by PMA or CLA, resulted in increased phosphorylation of pRb. Our results indicate that PP1 directly dephosphorylates pRb, while PP2A might have an indirect influence via mediating the phosphorylation level of PP1 inhibitory proteins. These data imply the importance of PP1 inhibitory proteins in controlling the phosphorylation state of key proteins and regulating drug sensitivity and apoptosis in leukemic cells.
Collapse
Affiliation(s)
- Dóra Dedinszki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Kiss
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Márkász
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Adrienn Márton
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Székely
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Ferenc Erdődi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
8
|
Prickaerts P, Niessen HE, Mouchel-Vielh E, Dahlmans VE, van den Akker GG, Geijselaers C, Adriaens ME, Spaapen F, Takihara Y, Rapp UR, Peronnet F, Voncken JW. MK3 controls Polycomb target gene expression via negative feedback on ERK. Epigenetics Chromatin 2012; 5:12. [PMID: 22870894 PMCID: PMC3499388 DOI: 10.1186/1756-8935-5-12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/11/2012] [Indexed: 01/04/2023] Open
Abstract
Background Gene-environment interactions are mediated by epigenetic mechanisms. Polycomb Group proteins constitute part of an epigenetic cellular transcriptional memory system that is subject to dynamic modulation during differentiation. Molecular insight in processes that control dynamic chromatin association and dissociation of Polycomb repressive complexes during and beyond development is limited. We recently showed that MK3 interacts with Polycomb repressive complex 1 (PRC1). The functional relevance of this interaction, however, remained poorly understood. MK3 is activated downstream of mitogen- and stress-activated protein kinases (M/SAPKs), all of which fulfill crucial roles during development. We here use activation of the immediate-early response gene ATF3, a bona fide PRC1 target gene, as a model to study how MK3 and its effector kinases MAPK/ERK and SAPK/P38 are involved in regulation of PRC1-dependent ATF3 transcription. Results Our current data show that mitogenic signaling through ERK, P38 and MK3 regulates ATF3 expression by PRC1/chromatin dissociation and epigenetic modulation. Mitogenic stimulation results in transient P38-dependent H3S28 phosphorylation and ERK-driven PRC1/chromatin dissociation at PRC1 targets. H3S28 phosphorylation by itself appears not sufficient to induce PRC1/chromatin dissociation, nor ATF3 transcription, as inhibition of MEK/ERK signaling blocks BMI1/chromatin dissociation and ATF3 expression, despite induced H3S28 phosphorylation. In addition, we establish that concomitant loss of local H3K27me3 promoter marking is not required for ATF3 activation. We identify pERK as a novel signaling-induced binding partner of PRC1, and provide evidence that MK3 controls ATF3 expression in cultured cells via negative regulatory feedback on M/SAPKs. Dramatically increased ectopic wing vein formation in the absence of Drosophila MK in a Drosophila ERK gain-of-function wing vein patterning model, supports the existence of MK-mediated negative feedback regulation on pERK. Conclusion We here identify and characterize important actors in a PRC1-dependent epigenetic signal/response mechanism, some of which appear to be nonspecific global responses, whereas others provide modular specificity. Our findings provide novel insight into a Polycomb-mediated epigenetic mechanism that dynamically controls gene transcription and support a direct link between PRC1 and cellular responses to changes in the microenvironment.
Collapse
Affiliation(s)
- Peggy Prickaerts
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands.,Laboratoire de Biologie du Développement UMR 7622, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie-Paris 6, 9 Quai Saint-Bernard, 75005, Paris, France
| | - Hanneke Ec Niessen
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| | - Emmanuèle Mouchel-Vielh
- Laboratoire de Biologie du Développement UMR 7622, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie-Paris 6, 9 Quai Saint-Bernard, 75005, Paris, France
| | - Vivian Eh Dahlmans
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| | - Guus Gh van den Akker
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| | - Claudia Geijselaers
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| | - Michiel E Adriaens
- BiGCaT Bioinformatics, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| | - Frank Spaapen
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| | - Yoshihiro Takihara
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Ulf R Rapp
- Department of Molecular Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Frédérique Peronnet
- Laboratoire de Biologie du Développement UMR 7622, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie-Paris 6, 9 Quai Saint-Bernard, 75005, Paris, France
| | - Jan Willem Voncken
- Department of Molecular Genetics, GROW School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 50, 6229ER, Maastricht, The Netherlands
| |
Collapse
|
9
|
Shen C, Huang Y, Liu Y, Wang G, Zhao Y, Wang Z, Teng M, Wang Y, Flockhart DA, Skaar TC, Yan P, Nephew KP, Huang THM, Li L. A modulated empirical Bayes model for identifying topological and temporal estrogen receptor α regulatory networks in breast cancer. BMC SYSTEMS BIOLOGY 2011; 5:67. [PMID: 21554733 PMCID: PMC3117732 DOI: 10.1186/1752-0509-5-67] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 05/09/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Estrogens regulate diverse physiological processes in various tissues through genomic and non-genomic mechanisms that result in activation or repression of gene expression. Transcription regulation upon estrogen stimulation is a critical biological process underlying the onset and progress of the majority of breast cancer. Dynamic gene expression changes have been shown to characterize the breast cancer cell response to estrogens, the every molecular mechanism of which is still not well understood. RESULTS We developed a modulated empirical Bayes model, and constructed a novel topological and temporal transcription factor (TF) regulatory network in MCF7 breast cancer cell line upon stimulation by 17β-estradiol stimulation. In the network, significant TF genomic hubs were identified including ER-alpha and AP-1; significant non-genomic hubs include ZFP161, TFDP1, NRF1, TFAP2A, EGR1, E2F1, and PITX2. Although the early and late networks were distinct (<5% overlap of ERα target genes between the 4 and 24 h time points), all nine hubs were significantly represented in both networks. In MCF7 cells with acquired resistance to tamoxifen, the ERα regulatory network was unresponsive to 17β-estradiol stimulation. The significant loss of hormone responsiveness was associated with marked epigenomic changes, including hyper- or hypo-methylation of promoter CpG islands and repressive histone methylations. CONCLUSIONS We identified a number of estrogen regulated target genes and established estrogen-regulated network that distinguishes the genomic and non-genomic actions of estrogen receptor. Many gene targets of this network were not active anymore in anti-estrogen resistant cell lines, possibly because their DNA methylation and histone acetylation patterns have changed.
Collapse
Affiliation(s)
- Changyu Shen
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yiwen Huang
- Division of Human Cancer Genetics, Ohio State University, Columbus, OH, 43210, USA
- Department of Molecular Virology, Immunology, and Medical Genetics, Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Yunlong Liu
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Guohua Wang
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Yuming Zhao
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Information and Computer Engineering College, Northeast Forestry University, Harbin, Heilongjiang, 150001, China
| | - Zhiping Wang
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mingxiang Teng
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - David A Flockhart
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Todd C Skaar
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pearlly Yan
- Division of Human Cancer Genetics, Ohio State University, Columbus, OH, 43210, USA
- Department of Molecular Virology, Immunology, and Medical Genetics, Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Kenneth P Nephew
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Departments of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Tim HM Huang
- Division of Human Cancer Genetics, Ohio State University, Columbus, OH, 43210, USA
- Department of Molecular Virology, Immunology, and Medical Genetics, Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Lang Li
- Center for Computational Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Sarver AL, Li L, Subramanian S. MicroRNA miR-183 functions as an oncogene by targeting the transcription factor EGR1 and promoting tumor cell migration. Cancer Res 2010; 70:9570-80. [PMID: 21118966 DOI: 10.1158/0008-5472.can-10-2074] [Citation(s) in RCA: 238] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transcription factor EGR1 is a tumor suppressor gene that is downregulated in many cancer types. Clinically, loss of EGR1 translates to increased tumor transformation and subsequent patient morbidity and mortality. In synovial sarcoma, the SS18-SSX fusion protein represses EGR1 expression through a direct association with the EGR1 promoter. However, the mechanism through which EGR1 becomes downregulated in other tumor types is unclear. Here, we report that EGR1 is regulated by microRNA (miR)-183 in multiple tumor types including synovial sarcoma, rhabdomyosarcoma (RMS), and colon cancer. Using an integrative network analysis, we identified that miR-183 is significantly overexpressed in these tumor types as well as in corresponding tumor cell lines. Bioinformatic analyses suggested that miR-183 could target EGR1 mRNA and this specific interaction was validated in vitro. miR-183 knockdown in synovial sarcoma, RMS, and colon cancer cell lines revealed deregulation of a miRNA network composed of miR-183-EGR1-PTEN in these tumors. Integrated miRNA- and mRNA-based genomic analyses indicated that miR-183 is an important contributor to cell migration in these tumor types and this result was functionally validated to be occurring via an EGR1-based mechanism. In conclusion, our findings have significant implications in the mechanisms underlying EGR1 regulation in cancers. miR-183 has a potential oncogenic role through the regulation of 2 tumor suppressor genes, EGR1 and PTEN, and the deregulation of this fundamental miRNA regulatory network may be central to many tumor types.
Collapse
Affiliation(s)
- Aaron L Sarver
- Biostatistics and Bioinformatics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
11
|
Eto M. Regulation of cellular protein phosphatase-1 (PP1) by phosphorylation of the CPI-17 family, C-kinase-activated PP1 inhibitors. J Biol Chem 2010; 284:35273-7. [PMID: 19846560 DOI: 10.1074/jbc.r109.059972] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The regulatory circuit controlling cellular protein phosphatase-1 (PP1), an abundant group of Ser/Thr phosphatases, involves phosphorylation of PP1-specific inhibitor proteins. Malfunctions of these inhibitor proteins have been linked to a variety of diseases, including cardiovascular disease and cancer. Upon phosphorylation at Thr(38), the 17-kDa PP1 inhibitor protein, CPI-17, selectively inhibits a specific form of PP1, myosin light chain phosphatase, which transduces multiple kinase signals into the phosphorylation of myosin II and other proteins. Here, the mechanisms underlying PP1 inhibition and the kinase/PP1 cross-talk mediated by CPI-17 and its related proteins, PHI, KEPI, and GBPI, are discussed.
Collapse
Affiliation(s)
- Masumi Eto
- Department of Molecular Physiology and Biophysics and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| |
Collapse
|
12
|
Zhu L, Johnson C, Bakovic M. Stimulation of the human CTP:phosphoethanolamine cytidylyltransferase gene by early growth response protein 1. J Lipid Res 2008; 49:2197-211. [PMID: 18583706 DOI: 10.1194/jlr.m800259-jlr200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Change in phosphoethanolamine pool size in tumor tissues is an important indicator of tumor prognosis and drug therapy efficacy. Phosphoethanolamine is the substrate of the regulatory enzyme CTP:phosphoethanolamine cytidylyltransferase (ECT) in the de novo biosynthesis of phosphatidylethanolamine (PE). Metabolic labeling with [14C]ethanolamine revealed a reduced ECT activity in MCF-7 breast cancer cells, which led to an accumulation of phosphoethanolamine and a decrease in PE synthesis in comparison with MCF-10A mammary epithelial cells. The enhanced ECT activity in MCF-10A cells was due to significantly elevated CTP:phosphoethanolamine cytidylyltransferase gene (PCYT2) expression, at the level of promoter activity, mRNA, and protein content. The early growth response protein 1 (EGR1) could account for most of the elevated ECT activity in MCF-10A cells relative to MCF-7 cells, as evidenced by promoter-luciferase reporter assays, gel-shift analyses, and by alterations in the EGR1 gene expression. In MCF-7 cells, EGR1 is present at lower levels and the basal PCYT2 promoter activity is maintained by proximal CAAT and GC regions and by elevated nuclear NFkappaB activity. Together, these data demonstrate that EGR1 is an important transcriptional stimulator of the human PCYT2 and that conditions that modify EGR1 also affect the function of ECT and consequently PE synthesis.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | |
Collapse
|