1
|
Wasserman JS, Faezov B, Patel KR, Kurimchak AM, Palacio SM, Glass DJ, Fowle H, McEwan BC, Xu Q, Zhao Z, Cressey L, Johnson N, Duncan JS, Kettenbach AN, Dunbrack RL, Graña X. FAM122A ensures cell cycle interphase progression and checkpoint control by inhibiting B55α/PP2A through helical motifs. Nat Commun 2024; 15:5776. [PMID: 38982062 PMCID: PMC11233601 DOI: 10.1038/s41467-024-50015-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
The Ser/Thr protein phosphatase 2 A (PP2A) regulates the dephosphorylation of many phosphoproteins. Substrate recognition are mediated by B regulatory subunits. Here, we report the identification of a substrate conserved motif [RK]-V-x-x-[VI]-R in FAM122A, an inhibitor of B55α/PP2A. This motif is necessary for FAM122A binding to B55α, and computational structure prediction suggests the motif, which is helical, blocks substrate docking to the same site. In this model, FAM122A also spatially constrains substrate access by occluding the catalytic subunit. Consistently, FAM122A functions as a competitive inhibitor as it prevents substrate binding and dephosphorylation of CDK substrates by B55α/PP2A in cell lysates. FAM122A deficiency in human cell lines reduces the proliferation rate, cell cycle progression, and hinders G1/S and intra-S phase cell cycle checkpoints. FAM122A-KO in HEK293 cells attenuates CHK1 and CHK2 activation in response to replication stress. Overall, these data strongly suggest that FAM122A is a short helical motif (SHeM)-dependent, substrate-competitive inhibitor of B55α/PP2A that suppresses multiple functions of B55α in the DNA damage response and in timely progression through the cell cycle interphase.
Collapse
Affiliation(s)
- Jason S Wasserman
- Fels Cancer Institute for Personalized Medicine. Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Bulat Faezov
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Kishan R Patel
- Fels Cancer Institute for Personalized Medicine. Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | | | - Seren M Palacio
- Fels Cancer Institute for Personalized Medicine. Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - David J Glass
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Holly Fowle
- Fels Cancer Institute for Personalized Medicine. Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Brennan C McEwan
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Medical Center Drive, Lebanon, NH, USA
| | - Qifang Xu
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Ziran Zhao
- Fels Cancer Institute for Personalized Medicine. Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Lauren Cressey
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Medical Center Drive, Lebanon, NH, USA
| | - Neil Johnson
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - James S Duncan
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Arminja N Kettenbach
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Medical Center Drive, Lebanon, NH, USA
| | | | - Xavier Graña
- Fels Cancer Institute for Personalized Medicine. Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA.
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Albalawi E. Genetic Rearrangements in Different Salivary Gland Tumors: A Systematic Review. Cureus 2024; 16:e61639. [PMID: 38966479 PMCID: PMC11223175 DOI: 10.7759/cureus.61639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/06/2024] Open
Abstract
Salivary gland tumors (SGT) encompass a wide range of neoplasms, each with its own unique histological type and clinical presentation. This review hones in on prevalent subtypes of SGTs, including adenoid cystic carcinoma (ACC), salivary duct carcinoma (SDC), and polymorphous adenocarcinoma (PAC). The articles, identified through specific keywords, were meticulously screened in databases like PubMed, Scopus, Google Scholar, and Web of Science from 2018 to 2023. Eight articles delved into genetic modifications among the selected SGT types. A fusion protein known as MYB-NF1B is typically associated with ACC, promoting cell proliferation while inhibiting apoptosis. The presence of MYB modifications in ACCs is a beacon of hope, as it is linked to a more favorable prognosis. In contrast, SDCs often exhibit HER2 expression. The invasive nature of SGTs contributes to their resistance to treatment. In the case of PAC, the role of PRKD1 is particularly noteworthy. PRKD1, integrated with other genes from the PRKD1/2/3 cluster, helps to differentiate PAC from other diseases. Furthermore, the genetic profiles of KTN1-PRKD1) and PPP2R2A:PRKD1 are distinct. The significant genetic variability among SGTs necessitates meticulous examination. This field is in a constant state of evolution, with new discoveries reshaping our understanding. Genetics is a key player in deciphering SGTs and tailoring treatments. This complex neoplasm demands ongoing research to uncover all genetic influences, thereby enhancing diagnostic methodologies, therapeutic strategies, and patient outcomes.
Collapse
|
3
|
de Jager VD, de Visscher SAHJ, Schuuring E, Doff JJ, van Kempen LC. A novel PPP2R2A::PRKD1 fusion in a cribriform adenocarcinoma of salivary gland. Genes Chromosomes Cancer 2023; 62:297-300. [PMID: 36625487 DOI: 10.1002/gcc.23122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/03/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023] Open
Abstract
Cribriform adenocarcinoma of salivary gland (CASG) is a rare, salivary gland tumor. In this report, we describe a case of CASG harboring a novel PPP2R2A::PRKD1 fusion. A 58-year-old female presented with an intraoral mass adjacent to the lower left third molar region. Morphological features at histological examination, immunohistochemical staining (p63+, p40-), and tumor location were indicative of CASG. However, due to the potential focal presence of a biphasic component within the tumor, RNA sequencing was performed to confirm the diagnosis. The subsequently found novel PPP2R2A::PRKD1 fusion adds to the rapidly evolving molecular landscape of salivary gland tumors. Additionally, we report that CASG may show some entrapment of pre-existent salivary gland ducts, which may be misinterpreted as tumor cells with myoepithelial differentiation.
Collapse
Affiliation(s)
- Vincent D de Jager
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sebastiaan A H J de Visscher
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan J Doff
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Léon C van Kempen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Greenberg ZJ, Paracatu LC, Monlish DA, Dong Q, Rettig M, Roundy N, Gaballa R, Li W, Yang W, Luke CJ, Schuettpelz LG. The tetraspanin CD53 protects stressed hematopoietic stem cells via promotion of DREAM complex-mediated quiescence. Blood 2023; 141:1180-1193. [PMID: 36542833 PMCID: PMC10023726 DOI: 10.1182/blood.2022016929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The hematopoietic stem cell (HSC) cycle responds to inflammatory and other proliferative stressors; however, these cells must quickly return to quiescence to avoid exhaustion and maintain their functional integrity. The mechanisms that regulate this return to quiescence are not well understood. Here, we show that tetraspanin CD53 is markedly upregulated in HSCs in response to a variety of inflammatory and proliferative stimuli and that the loss of CD53 is associated with prolonged cycling and reduced HSC function in the context of inflammatory stress. Mechanistically, CD53 promotes the activity of the dimerization partner, RB-like, E2F, and multi-vulva class B (DREAM) transcriptional repressor complex, which downregulates genes associated with cycling and division. Proximity labeling and confocal fluorescence microscopy studies showed that CD53 interacts with DREAM-associated proteins, specifically promoting the interaction between Rbl2/p130 and its phosphatase protein phosphatase 2A (PP2A), effectively stabilizing p130 protein availability for DREAM binding. Together, these data identified a novel mechanism by which stressed HSCs resist cycling.
Collapse
Affiliation(s)
- Zev J. Greenberg
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | | | - Darlene A. Monlish
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Qian Dong
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Michael Rettig
- Department of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Nate Roundy
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Rofaida Gaballa
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Weikai Li
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Cliff J. Luke
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Laura G. Schuettpelz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
5
|
Shao L, Ma Y, Fang Q, Huang Z, Wan S, Wang J, Yang L. Role of protein phosphatase 2A in kidney disease (Review). Exp Ther Med 2021; 22:1236. [PMID: 34539832 PMCID: PMC8438693 DOI: 10.3892/etm.2021.10671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Kidney disease affects millions of people worldwide and is a financial burden on the healthcare system. Protein phosphatase 2A (PP2A), which is involved in renal development and the function of ion-transport proteins, aquaporin-2 and podocytes, is likely to serve an important role in renal processes. PP2A is associated with the pathogenesis of a variety of different kidney diseases including podocyte injury, inflammation, tumors and chronic kidney disease. The current review aimed to discuss the structure and function of PP2A subunits in the context of kidney diseases. How dysregulation of PP2A in the kidneys causes podocyte death and the inactivation of PP2A in renal carcinoma tissues is discussed. Inhibition of PP2A activity prevents epithelial-mesenchymal transition and attenuates renal fibrosis, creating a favorable inflammatory microenvironment and promoting the initiation and progression of tumor pathogenesis. The current review also indicates that PP2A serves an important role in protection against renal inflammation. Understanding the detailed mechanisms of PP2A provides information that can be utilized in the design and application of novel therapeutics for the treatment and prevention of renal diseases.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Yiqun Ma
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| | - Ziye Huang
- Department of Urology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Shanshan Wan
- Department of Radiology, Yunnan Kun-Gang Hospital, Anning, Yunnan 650300, P.R. China
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Li Yang
- Department of Anatomy, Histology and Embryology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
6
|
Bao Y, Oguz G, Lee WC, Lee PL, Ghosh K, Li J, Wang P, Lobie PE, Ehmsen S, Ditzel HJ, Wong A, Tan EY, Lee SC, Yu Q. EZH2-mediated PP2A inactivation confers resistance to HER2-targeted breast cancer therapy. Nat Commun 2020; 11:5878. [PMID: 33208750 PMCID: PMC7674491 DOI: 10.1038/s41467-020-19704-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
HER2-targeted therapy has yielded a significant clinical benefit in patients with HER2+ breast cancer, yet disease relapse due to intrinsic or acquired resistance remains a significant challenge in the clinic. Here, we show that the protein phosphatase 2A (PP2A) regulatory subunit PPP2R2B is a crucial determinant of anti-HER2 response. PPP2R2B is downregulated in a substantial subset of HER2+ breast cancers, which correlates with poor clinical outcome and resistance to HER2-targeted therapies. EZH2-mediated histone modification accounts for the PPP2R2B downregulation, resulting in sustained phosphorylation of PP2A targets p70S6K and 4EBP1 which leads to resistance to inhibition by anti-HER2 treatments. Genetic depletion or inhibition of EZH2 by a clinically-available EZH2 inhibitor restores PPP2R2B expression, abolishes the residual phosphorylation of p70S6K and 4EBP1, and resensitizes HER2+ breast cancer cells to anti-HER2 treatments both in vitro and in vivo. Furthermore, the same epigenetic mechanism also contributes to the development of acquired resistance through clonal selection. These findings identify EZH2-dependent PPP2R2B suppression as an epigenetic control of anti-HER2 resistance, potentially providing an opportunity to mitigate anti-HER2 resistance with EZH2 inhibitors.
Collapse
Affiliation(s)
- Yi Bao
- Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Cancer Precision Medicine, Genome Institute of Singapore, Agency for Science, Technology, and Research, Biopolis, Singapore, 138672, Singapore
| | - Gokce Oguz
- Cancer Precision Medicine, Genome Institute of Singapore, Agency for Science, Technology, and Research, Biopolis, Singapore, 138672, Singapore
| | - Wee Chyan Lee
- Cancer Precision Medicine, Genome Institute of Singapore, Agency for Science, Technology, and Research, Biopolis, Singapore, 138672, Singapore
| | - Puay Leng Lee
- Cancer Precision Medicine, Genome Institute of Singapore, Agency for Science, Technology, and Research, Biopolis, Singapore, 138672, Singapore
| | - Kakaly Ghosh
- Cancer Precision Medicine, Genome Institute of Singapore, Agency for Science, Technology, and Research, Biopolis, Singapore, 138672, Singapore
| | - Jiayao Li
- Cancer Research Institute, Jinan University, Guangzhou, China
| | - Panpan Wang
- Cancer Research Institute, Jinan University, Guangzhou, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,Tsinghua-Berkeley Shenzhen Institute, Guangdong Province and Shenzhen Bay Laboratory, Tsinghua University, Shenzhen, Guangdong Province, China
| | - Sidse Ehmsen
- Department of Oncology, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, 5230, Odense, Denmark
| | - Henrik J Ditzel
- Department of Oncology, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, 5230, Odense, Denmark.,Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5230, Odense, Denmark
| | - Andrea Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, 119047, Singapore
| | - Ern Yu Tan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Soo Chin Lee
- Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore. .,Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, 119047, Singapore.
| | - Qiang Yu
- Cancer Precision Medicine, Genome Institute of Singapore, Agency for Science, Technology, and Research, Biopolis, Singapore, 138672, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore. .,Cancer and Stem Cell Biology, DUKE-NUS Graduate Medical School of Singapore, Singapore, 169857, Singapore.
| |
Collapse
|
7
|
MnTE-2-PyP Suppresses Prostate Cancer Cell Growth via H 2O 2 Production. Antioxidants (Basel) 2020; 9:antiox9060490. [PMID: 32512786 PMCID: PMC7346125 DOI: 10.3390/antiox9060490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/03/2020] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer patients are often treated with radiotherapy. MnTE-2-PyP, a superoxide dismutase (SOD) mimic, is a known radioprotector of normal tissues. Our recent work demonstrated that MnTE-2-PyP also inhibits prostate cancer progression with radiotherapy; however, the mechanisms remain unclear. In this study, we identified that MnTE-2-PyP-induced intracellular H2O2 levels are critical in inhibiting the growth of PC3 and LNCaP cells, but the increased H2O2 levels affected the two cancer cells differently. In PC3 cells, many proteins were thiol oxidized with MnTE-2-PyP treatment, including Ser/Thr protein phosphatase 1 beta catalytic subunit (PP1CB). This resulted in reduced PP1CB activity; however, overall cell cycle progression was not altered, so this is not the main mechanism of PC3 cell growth inhibition. High H2O2 levels by MnTE-2-PyP treatment induced nuclear fragmentation, which could be synergistically enhanced with radiotherapy. In LNCaP cells, thiol oxidation by MnTE-2-PyP treatment was not observed previously and, similarly to PC3 cells, there was no effect of MnTE-2-PyP treatment on cell cycle progression. However, in LNCaP cells, MnTE-2-PyP caused an increase in low RNA population and sub-G1 population of cells, which indicates that MnTE-2-PyP treatment may cause cellular quiescence or direct cancer cell death. The protein oxidative modifications and mitotic catastrophes caused by MnTE-2-PyP may be the major contributors to cell growth inhibition in PC3 cells, while in LNCaP cells, tumor cell quiescence or cell death appears to be major factors in MnTE-2-PyP-induced growth inhibition.
Collapse
|
8
|
Zhao Z, Kurimchak A, Nikonova AS, Feiser F, Wasserman JS, Fowle H, Varughese T, Connors M, Johnson K, Makhov P, Lindskog C, Kolenko VM, Golemis EA, Duncan JS, Graña X. PPP2R2A prostate cancer haploinsufficiency is associated with worse prognosis and a high vulnerability to B55α/PP2A reconstitution that triggers centrosome destabilization. Oncogenesis 2019; 8:72. [PMID: 31822657 PMCID: PMC6904742 DOI: 10.1038/s41389-019-0180-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
The PPP2R2A gene encodes the B55α regulatory subunit of PP2A. Here, we report that PPP2R2A is hemizygously lost in ~42% of prostate adenocarcinomas, correlating with reduced expression, poorer prognosis, and an increased incidence of hemizygous loss (>75%) in metastatic disease. Of note, PPP2R2A homozygous loss is less common (5%) and not increased at later tumor stages. Reduced expression of B55α is also seen in prostate tumor tissue and cell lines. Consistent with the possibility that complete loss of PPP2R2A is detrimental in prostate tumors, PPP2R2A deletion in cells with reduced but present B55α reduces cell proliferation by slowing progression through the cell cycle. Remarkably, B55α-low cells also appear addicted to lower B55α expression, as even moderate increases in B55α expression are toxic. Reconstitution of B55α expression in prostate cancer (PCa) cell lines with low B55α expression reduces proliferation, inhibits transformation and blocks xenograft tumorigenicity. Mechanistically, we show B55α reconstitution reduces phosphorylation of proteins essential for centrosomal maintenance, and induces centrosome collapse and chromosome segregation failure; a first reported link between B55α/PP2A and the vertebrate centrosome. These effects are dependent on a prolonged metaphase/anaphase checkpoint and are lethal to PCa cells addicted to low levels of B55α. Thus, we propose the reduction in B55α levels associated with hemizygous loss is necessary for centrosomal integrity in PCa cells, leading to selective lethality of B55α reconstitution. Such a vulnerability could be targeted therapeutically in the large pool of patients with hemizygous PPP2R2A deletions, using pharmacologic approaches that enhance PP2A/B55α activity.
Collapse
Affiliation(s)
- Ziran Zhao
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Alison Kurimchak
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.,Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | - Felicity Feiser
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Jason S Wasserman
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Holly Fowle
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Tinsa Varughese
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Megan Connors
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | | | - Petr Makhov
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 36, Uppsala, Sweden
| | | | | | | | - Xavier Graña
- Fels Institute for Cancer Research and Molecular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
9
|
Fowle H, Zhao Z, Graña X. PP2A holoenzymes, substrate specificity driving cellular functions and deregulation in cancer. Adv Cancer Res 2019; 144:55-93. [PMID: 31349904 PMCID: PMC9994639 DOI: 10.1016/bs.acr.2019.03.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PP2A is a highly conserved eukaryotic serine/threonine protein phosphatase of the PPP family of phosphatases with fundamental cellular functions. In cells, PP2A targets specific subcellular locations and substrates by forming heterotrimeric holoenzymes, where a core dimer consisting of scaffold (A) and catalytic (C) subunits complexes with one of many B regulatory subunits. PP2A plays a key role in positively and negatively regulating a myriad of cellular processes, as it targets a very sizable fraction of the cellular substrates phosphorylated on Ser/Thr residues. This review focuses on insights made toward the understanding on how the subunit composition and structure of PP2A holoenzymes mediates substrate specificity, the role of substrate modulation in the signaling of cellular division, growth, and differentiation, and its deregulation in cancer.
Collapse
Affiliation(s)
- Holly Fowle
- Fels Institute for Cancer Research and Molecular Biology and Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Ziran Zhao
- Fels Institute for Cancer Research and Molecular Biology and Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xavier Graña
- Fels Institute for Cancer Research and Molecular Biology and Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
10
|
Yoon JT, Ahn HK, Pai HS. The subfamily II catalytic subunits of protein phosphatase 2A (PP2A) are involved in cortical microtubule organization. PLANTA 2018; 248:1551-1567. [PMID: 30191298 DOI: 10.1007/s00425-018-3000-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 08/30/2018] [Indexed: 05/07/2023]
Abstract
The subfamily II catalytic subunits of protein phosphatase 2A (PP2A) regulate the cortical microtubule dynamics in Arabidopsis, through interaction with TONNEAU2 (TON2)/FASS and modulation of α-tubulin dephosphorylation. Protein phosphatase 2A is a major protein phosphatase in eukaryotes that dephosphorylates many different substrates to regulate their function. PP2A is assembled into a heterotrimeric complex of scaffolding A subunit, regulatory B subunit, and catalytic C subunit. Plant PP2A catalytic C subunit (PP2AC) isoforms are classified into two subfamilies. In this study, we investigated the cellular functions of the Arabidopsis PP2AC subfamily II genes PP2AC-3 and PP2AC-4, particularly regarding the cortical microtubule (MT) organization. PP2AC-3 and PP2AC-4 strongly interacted with the B'' regulatory subunit TON2. Simultaneous silencing of PP2AC-3 and PP2AC-4 by virus-induced gene silencing (PP2AC-3,4 VIGS) significantly altered plant morphology in Arabidopsis, increasing cell numbers in leaves and stems. The leaf epidermis of PP2AC-3,4 VIGS plants largely lost its jigsaw-puzzle shape and exhibited reduced trichome branch numbers. VIGS of PP2AC-3,4 in Arabidopsis transgenic plants that expressed GFP-fused β-tubulin 6 isoform (GFP-TUB6) for the visualization of MTs caused a reduction in the cortical MT array density in the pavement cells. VIGS of TON2 also led to similar cellular phenotypes and cortical MT patterns compared with those after VIGS of PP2AC-3,4, suggesting that PP2AC-3,4 and their interaction partner TON2 play a role in cortical MT organization in leaf epidermal cells. Furthermore, silencing of PP2AC-3,4 did not affect salt-induced phosphorylation of α-tubulin but delayed its dephosphorylation after salt removal. The reappearance of cortical MT arrays after salt removal was impaired in PP2AC-3,4 VIGS plants. These results suggest an involvement of PP2AC subfamily II in the regulation of cortical MT dynamics under normal and salt-stress conditions in Arabidopsis.
Collapse
Affiliation(s)
- Joong-Tak Yoon
- Department of Systems Biology, Yonsei University, Seoul, 03722, Korea
| | - Hee-Kyung Ahn
- Department of Systems Biology, Yonsei University, Seoul, 03722, Korea
- The Sainsbury Laboratory (TSL), Norwich Research Park, Norwich, NR4 7UH, UK
| | - Hyun-Sook Pai
- Department of Systems Biology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
11
|
Liang S, Guo J, Choi JW, Shin KT, Wang HY, Jo YJ, Kim NH, Cui XS. Protein phosphatase 2A regulatory subunit B55α functions in mouse oocyte maturation and early embryonic development. Oncotarget 2018; 8:26979-26991. [PMID: 28439046 PMCID: PMC5432312 DOI: 10.18632/oncotarget.15927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Protein phosphatase 2A regulatory subunit B55α (PP2A-B55α) has been studied in mitosis. However, its functions in mammalian meiosis and early embryonic development remain unknown. Here, we report that PP2A-B55α is critical for mouse oocyte meiosis and preimplantation embryo development. Knockdown of PP2A-B55α in oocytes led to abnormal asymmetric division, disordered spindle dynamics, defects in chromosome congression, an increase in aneuploidy, and induction of the DNA damage response. Moreover, knockdown of PP2A-B55α in fertilized mouse zygotes impaired development to the blastocyst stage. The impairment of embryonic development might have been due to induction of sustained DNA damage in embryos, which caused apoptosis and inhibited cell proliferation and outgrowth potential at the blastocyst stage. Overall, these results provide a novel insight into the role of PP2A-B55α as a novel meiotic and embryonic competence factor at the onset of life.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Jing Guo
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Jeong-Woo Choi
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Kyung-Tae Shin
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Hai-Yang Wang
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Yu-Jin Jo
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Nam-Hyung Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| |
Collapse
|
12
|
PP2A inhibitors arrest G2/M transition through JNK/Sp1- dependent down-regulation of CDK1 and autophagy-dependent up-regulation of p21. Oncotarget 2016; 6:18469-83. [PMID: 26053095 PMCID: PMC4621904 DOI: 10.18632/oncotarget.4063] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/14/2015] [Indexed: 01/07/2023] Open
Abstract
Protein phosphatase 2A (PP2A) plays an important role in the control of the cell cycle. We previously reported that the PP2A inhibitors, cantharidin and okadaic acid (OA), efficiently repressed the growth of cancer cells. In the present study, we found that PP2A inhibitors arrested the cell cycle at the G2 phase through a mechanism that was dependent on the JNK pathway. Microarrays further showed that PP2A inhibitors induced expression changes in multiple genes that participate in cell cycle transition. To verify whether these expression changes were executed in a PP2A-dependent manner, we targeted the PP2A catalytic subunit (PP2Ac) using siRNA and evaluated gene expression with a microarray. After the cross comparison of these microarray data, we identified that CDK1 was potentially the same target when treated with either PP2A inhibitors or PP2Ac siRNA. In addition, we found that the down-regulation of CDK1 occurred in a JNK-dependent manner. Luciferase reporter gene assays demonstrated that repression of the transcription of CDK1 was executed through the JNK-dependent activation of the Sp1 transcription factor. By constructing deletion mutants of the CDK1 promoter and by using ChIP assays, we identified an element in the CDK1 promoter that responded to the JNK/Sp1 pathway after stimulation with PP2A inhibitors. Cantharidin and OA also up-regulated the expression of p21, an inhibitor of CDK1, via autophagy rather than PP2A/JNK pathway. Thus, this present study found that the PP2A/JNK/Sp1/CDK1 pathway and the autophagy/p21 pathway participated in G2/M cell cycle arrest triggered by PP2A inhibitors.
Collapse
|
13
|
Aldiri I, Ajioka I, Xu B, Zhang J, Chen X, Benavente C, Finkelstein D, Johnson D, Akiyama J, Pennacchio LA, Dyer MA. Brg1 coordinates multiple processes during retinogenesis and is a tumor suppressor in retinoblastoma. Development 2016; 142:4092-106. [PMID: 26628093 PMCID: PMC4712833 DOI: 10.1242/dev.124800] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Retinal development requires precise temporal and spatial coordination of cell cycle exit, cell fate specification, cell migration and differentiation. When this process is disrupted, retinoblastoma, a developmental tumor of the retina, can form. Epigenetic modulators are central to precisely coordinating developmental events, and many epigenetic processes have been implicated in cancer. Studying epigenetic mechanisms in development is challenging because they often regulate multiple cellular processes; therefore, elucidating the primary molecular mechanisms involved can be difficult. Here we explore the role of Brg1 (Smarca4) in retinal development and retinoblastoma in mice using molecular and cellular approaches. Brg1 was found to regulate retinal size by controlling cell cycle length, cell cycle exit and cell survival during development. Brg1 was not required for cell fate specification but was required for photoreceptor differentiation and cell adhesion/polarity programs that contribute to proper retinal lamination during development. The combination of defective cell differentiation and lamination led to retinal degeneration in Brg1-deficient retinae. Despite the hypocellularity, premature cell cycle exit, increased cell death and extended cell cycle length, retinal progenitor cells persisted in Brg1-deficient retinae, making them more susceptible to retinoblastoma. ChIP-Seq analysis suggests that Brg1 might regulate gene expression through multiple mechanisms. Summary: The SWI/SNF protein Brg1 controls cell cycle length, cell cycle exit and cell survival, and is required for cell differentiation and retinal lamination, in the developing mouse retina.
Collapse
Affiliation(s)
- Issam Aldiri
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Itsuki Ajioka
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiakun Zhang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Claudia Benavente
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dianna Johnson
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer Akiyama
- Lawrence Berkeley National Laboratory, Genomics Division, Berkeley, CA 94701, USA Department of Energy, Joint Genome Institute, Walnut Creek, CA 94598, USA
| | - Len A Pennacchio
- Lawrence Berkeley National Laboratory, Genomics Division, Berkeley, CA 94701, USA Department of Energy, Joint Genome Institute, Walnut Creek, CA 94598, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
14
|
Nagel AC, Szawinski J, Zimmermann M, Preiss A. Drosophila Cyclin G Is a Regulator of the Notch Signalling Pathway during Wing Development. PLoS One 2016; 11:e0151477. [PMID: 26963612 PMCID: PMC4786218 DOI: 10.1371/journal.pone.0151477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/29/2016] [Indexed: 01/24/2023] Open
Abstract
Notch signalling regulates a multitude of differentiation processes during Drosophila development. For example, Notch activity is required for proper wing vein differentiation which is hampered in mutants of either the receptor Notch, the ligand Delta or the antagonist Hairless. Moreover, the Notch pathway is involved in several aspects of Drosophila oogenesis as well. We have identified Drosophila Cyclin G (CycG) as a molecular interaction partner of Hairless, the major antagonist in the Notch signalling pathway, in vitro and in vivo. Loss of CycG was shown before to cause female sterility and to disturb the architecture of the egg shell. Nevertheless, Notch dependent processes during oogenesis appeared largely unaffected in cycG mutant egg chambers. Loss of CycG modified the dominant wing phenotypes of Notch, Delta and Hairless mutants. Whereas the Notch loss of function phenotype was ameliorated by a loss of CycG, the phenotypes of either Notch gain of function or of Delta or Hairless loss of function were enhanced. In contrast, loss of CycG had only a minor effect on the wing vein phenotype of mutants affecting the EGFR signalling pathway emphasizing the specificity of the interaction of CycG and Notch pathway members.
Collapse
Affiliation(s)
- Anja C. Nagel
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Jutta Szawinski
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Mirjam Zimmermann
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anette Preiss
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
- * E-mail:
| |
Collapse
|
15
|
Zhang W, Chen H, Chen Y, Liu J, Wang X, Yu X, Chen JJ, Zhao W. Cancerous inhibitor of protein phosphatase 2A contributes to human papillomavirus oncoprotein E7-induced cell proliferation via E2F1. Oncotarget 2016; 6:5253-62. [PMID: 25650660 PMCID: PMC4467146 DOI: 10.18632/oncotarget.2867] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 12/07/2014] [Indexed: 12/31/2022] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that is overexpressed in many human malignant tumors including cervical cancer. Human papillomavirus (HPV) oncoprotein E7 is the key transformation factor in cervical cancer. Our previous data showed a positive association of CIP2A and HPV-16E7 protein levels; however, how CIP2A is regulated by HPV-E7 and the roles of CIP2A in HPV-E7-mediated cell proliferation are unknown. In this study, we demonstrated that HPV-16E7 protein significantly upregulating CIP2A mRNA and protein expression depended on retinoblastoma protein pRb rather than p130. CIP2A siRNA knockdown in HPV-E7-expressing cells inhibited cell proliferation, DNA synthesis and G1/S cell cycle progression. CIP2A siRNA decreased the protein levels of cyclin-dependent kinase 1 (Cdk1), Cdk2 and their partner cyclin A2, with no change in levels of Cdk4, Cdk6 and their partner cyclin D1. The downregulation of Cdk1 and Cdk2 was independent of c-Myc; instead, E2F1 was the main target of CIP2A in this process, as overexpression of E2F1 rescued the inhibitory effects of CIP2A siRNA knockdown on cell proliferation and G1 arrest of HPV-E7-expressing cells. Our studies reveal a novel function of CIP2A in HPV-16E7-mediated cell proliferation.
Collapse
Affiliation(s)
- Weifang Zhang
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Hanxiang Chen
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Yan Chen
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Juan Liu
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xiao Wang
- Institute of Pathophysiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xiuping Yu
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Jason J Chen
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,Cancer Research Center, Shandong University School of Medicine, Jinan, Shandong, China
| | - Weiming Zhao
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong, China
| |
Collapse
|
16
|
Kurimchak A, Graña X. PP2A: more than a reset switch to activate pRB proteins during the cell cycle and in response to signaling cues. Cell Cycle 2015; 14:18-30. [PMID: 25483052 PMCID: PMC4612414 DOI: 10.4161/15384101.2014.985069] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In their active hypophosphorylated state, members of the retinoblastoma family of pocket proteins negatively regulate cell cycle progression at least in part by repressing expression of E2F-dependent genes. Mitogen-dependent activation of G1 and G1/S Cyclin Dependent Kinases (CDKs) results in coordinated hyperphosphorylation and inactivation of these proteins, which no longer bind and repress E2Fs. S and G2/M CDKs maintain pocket protein hyperphosphorylated through the end of mitosis. The inactivating action of inducible CDKs is opposed by the Ser/Thr protein phosphatases PP2A and PP1. Various trimeric PP2A holoenzymes have been implicated in dephosphorylation of pocket proteins in response to specific cellular signals and stresses or as part of an equilibrium with CDKs throughout the cell cycle. PP1 has specifically been implicated in dephosphorylation of pRB in late mitosis and early G1. This review is particularly focused on the emerging role of PP2A as a major hub for integration of growth suppressor signals that require rapid inactivation of pocket proteins. Of note, activation of particular PP2A holoenzymes triggers differential activation of pocket proteins in the presence of active CDKs.
Collapse
Affiliation(s)
- Alison Kurimchak
- a Fels Institute for Cancer Research and Molecular Biology and Department of Biochemistry; Temple University School of Medicine ; Philadelphia , PA USA
| | | |
Collapse
|
17
|
Zhao W, Cao L, Zeng S, Qin H, Men T. Upregulation of miR-556-5p promoted prostate cancer cell proliferation by suppressing PPP2R2A expression. Biomed Pharmacother 2015; 75:142-7. [PMID: 26297546 DOI: 10.1016/j.biopha.2015.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 07/26/2015] [Indexed: 12/12/2022] Open
Abstract
The prognosis and survival rate of prostate cancer are very poor. Previous studies have shown that miR-556-5p have emerged as important regulators in cancer cell biological processes. The role of miR-556-5p in prostate cancer remains unclear. In this study, expression of miR-556-5p in prostate cancer cell lines and tissues was upregulated. Result of MTT assays, colony formation and anchorage-independent growth assays demonstrated that overexpression of miR-556-5p promoted prostate cancer cell growth. Additionally, PPP2R2A was identified as a direct target of miR-556-5p. Ectopic expression of miR-556-5p led to downregulation of PPP2R2A protein, which resulted in the downregulation of p27, upregulation of cyclin D1. Taken together, our data provide compelling evidence that miR-556-5p functions as an onco-miRNA and participates in prostate cancer carcinogenesis by suppressing PPP2R2A expression.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Urology, Qianfoshan Hospital Affiliated to Shandong University, Jinan 250014, Shandong, China; Department of Urology, Liaocheng People's Hospital, Liaocheng 252000, Shandong, China
| | - Longbin Cao
- Department of Urology, Liaocheng People's Hospital, Liaocheng 252000, Shandong, China
| | - Sha Zeng
- Central Laboratory, Liaocheng People's Hospital, Liaocheng 252000, Shandong, China
| | - Haiping Qin
- Central Laboratory, Liaocheng People's Hospital, Liaocheng 252000, Shandong, China
| | - Tongyi Men
- Department of Urology, Qianfoshan Hospital Affiliated to Shandong University, Jinan 250014, Shandong, China.
| |
Collapse
|
18
|
Fischer P, La Rosa MK, Schulz A, Preiss A, Nagel AC. Cyclin G Functions as a Positive Regulator of Growth and Metabolism in Drosophila. PLoS Genet 2015; 11:e1005440. [PMID: 26274446 PMCID: PMC4537266 DOI: 10.1371/journal.pgen.1005440] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 07/13/2015] [Indexed: 01/15/2023] Open
Abstract
In multicellular organisms, growth and proliferation is adjusted to nutritional conditions by a complex signaling network. The Insulin receptor/target of rapamycin (InR/TOR) signaling cascade plays a pivotal role in nutrient dependent growth regulation in Drosophila and mammals alike. Here we identify Cyclin G (CycG) as a regulator of growth and metabolism in Drosophila. CycG mutants have a reduced body size and weight and show signs of starvation accompanied by a disturbed fat metabolism. InR/TOR signaling activity is impaired in cycG mutants, combined with a reduced phosphorylation status of the kinase Akt1 and the downstream factors S6-kinase and eukaryotic translation initiation factor 4E binding protein (4E-BP). Moreover, the expression and accumulation of Drosophila insulin like peptides (dILPs) is disturbed in cycG mutant brains. Using a reporter assay, we show that the activity of one of the first effectors of InR signaling, Phosphoinositide 3-kinase (PI3K92E), is unaffected in cycG mutants. However, the metabolic defects and weight loss in cycG mutants were rescued by overexpression of Akt1 specifically in the fat body and by mutants in widerborst (wdb), the B'-subunit of the phosphatase PP2A, known to downregulate Akt1 by dephosphorylation. Together, our data suggest that CycG acts at the level of Akt1 to regulate growth and metabolism via PP2A in Drosophila. Size and growth of an organism are adjusted to nutritional conditions by a complex regulatory network involving the Insulin receptor and TOR signaling cascades. Drosophila melanogaster has been used in the past as a genetically tractable model to unravel the complex circuitry by genetic means. We have identified CycG as an important player in the regulation of TOR signaling. CycG mutants are underweight in the midst of food and show typical signs of TOR defects. We provide evidence that CycG acts at the level of Akt1 kinase that links the Insulin receptor and TOR signaling cascades. Molecular and genetic data point to an interplay of CycG and phosphatase PP2A, a well established negative regulator of Akt1 activity. Moreover, CycG may influence PP2A-Akt1 binding. We propose that CycG, by impeding PP2A-Akt1 interaction, acts as a positive regulator of growth in Drosophila.
Collapse
Affiliation(s)
- Patrick Fischer
- Institute of Genetics, University of Hohenheim, Stuttgart, Germany
| | | | - Adriana Schulz
- Institute of Genetics, University of Hohenheim, Stuttgart, Germany
| | - Anette Preiss
- Institute of Genetics, University of Hohenheim, Stuttgart, Germany
| | - Anja C. Nagel
- Institute of Genetics, University of Hohenheim, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
19
|
Sun D, Buttitta L. Protein phosphatase 2A promotes the transition to G0 during terminal differentiation in Drosophila. Development 2015; 142:3033-45. [PMID: 26253406 DOI: 10.1242/dev.120824] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 07/28/2015] [Indexed: 12/22/2022]
Abstract
Protein phosphatase type 2A complex (PP2A) has been known as a tumor suppressor for over two decades, but it remains unclear exactly how it suppresses tumor growth. Here, we provide data indicating a novel role for PP2A in promoting the transition to quiescence upon terminal differentiation in vivo. Using Drosophila eyes and wings as a model, we find that compromising PP2A activity during the final cell cycle prior to a developmentally controlled cell cycle exit leads to extra cell divisions and delays entry into quiescence. By systematically testing the regulatory subunits of Drosophila PP2A, we find that the B56 family member widerborst (wdb) is required for the role of PP2A in promoting the transition to quiescence. Cells in differentiating tissues with compromised PP2A retain high Cdk2 activity when they should be quiescent, and genetic epistasis tests demonstrate that ectopic Cyclin E/Cdk2 activity is responsible for the extra cell cycles caused by PP2A inhibition. The loss of wdb/PP2A function cooperates with aberrantly high Cyclin E protein levels, allowing cells to bypass a robust G0 late in development. This provides an example of how loss of PP2A can cooperate with oncogenic mutations in cancer. We propose that the PP2A complex plays a novel role in differentiating tissues to promote developmentally controlled quiescence through the regulation of Cyclin E/Cdk2 activity.
Collapse
Affiliation(s)
- Dan Sun
- University of Michigan, Department of Molecular, Cellular and Developmental Biology, Ann Arbor, MI 48109, USA
| | - Laura Buttitta
- University of Michigan, Department of Molecular, Cellular and Developmental Biology, Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
Kiely M, Kiely PA. PP2A: The Wolf in Sheep's Clothing? Cancers (Basel) 2015; 7:648-69. [PMID: 25867001 PMCID: PMC4491676 DOI: 10.3390/cancers7020648] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/16/2015] [Accepted: 03/23/2015] [Indexed: 12/21/2022] Open
Abstract
Protein Phosphatase 2A (PP2A) is a major serine/threonine phosphatase in cells. It consists of a catalytic subunit (C), a structural subunit (A), and a regulatory/variable B-type subunit. PP2A has a critical role to play in homeostasis where its predominant function is as a phosphatase that regulates the major cell signaling pathways in cells. Changes in the assembly, activity and substrate specificity of the PP2A holoenzyme have a direct role in disease and are a major contributor to the maintenance of the transformed phenotype in cancer. We have learned a lot about how PP2A functions from specific mutations that disrupt the core assembly of PP2A and from viral proteins that target PP2A and inhibit its effect as a phosphatase. This prompted various studies revealing that restoration of PP2A activity benefits some cancer patients. However, our understanding of the mechanism of action of this is limited because of the complex nature of PP2A holoenzyme assembly and because it acts through a wide variety of signaling pathways. Information on PP2A is also conflicting as there are situations whereby inactivation of PP2A induces apoptosis in many cancer cells. In this review we discuss this relationship and we also address many of the pertinent and topical questions that relate to novel therapeutic strategies aimed at altering PP2A activity.
Collapse
Affiliation(s)
- Maeve Kiely
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick 78666, Ireland.
| | - Patrick A Kiely
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick 78666, Ireland.
- Stokes Institute, University of Limerick 78666, Limerick, Ireland.
| |
Collapse
|
21
|
miR-136 modulates TGF-β1-induced proliferation arrest by targeting PPP2R2A in keratinocytes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:453518. [PMID: 25654102 PMCID: PMC4310454 DOI: 10.1155/2015/453518] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 09/23/2014] [Indexed: 12/22/2022]
Abstract
Keratinocytes proliferation is critical for the capacity to heal wounds and accumulating evidences have proved that dysregulation of microRNAs is involved in proliferation of keratinocytes. However, the molecular mechanisms remain to be completely elucidated. Here, we show that miR-136 was significantly decreased by TGF-β1 treatment in HaCaT cells and normal human epidermal keratinocytes (NHEK), and it was a Smad3-dependent manner. By cell proliferation assay and cell cycle analysis, we found that reintroduction of miR-136 by transfection, as well as PPP2R2A silencing, counteracted TGF-β-induced proliferation arrest in HaCaT cells. Further, PPP2R2A was verified as a direct target of miR-136 by dual-luciferase reporter assays and Western blotting. These data suggest that miR-136 may play an important role during TGF-β1-induced proliferation arrest by targeting PPP2R2A in keratinocytes, which might represent a potential target for improving skin wound healing.
Collapse
|
22
|
Nguyen TA, Menendez D, Resnick MA, Anderson CW. Mutant TP53 posttranslational modifications: challenges and opportunities. Hum Mutat 2014; 35:738-55. [PMID: 24395704 DOI: 10.1002/humu.22506] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022]
Abstract
The wild-type (WT) human p53 (TP53) tumor suppressor can be posttranslationally modified at over 60 of its 393 residues. These modifications contribute to changes in TP53 stability and in its activity as a transcription factor in response to a wide variety of intrinsic and extrinsic stresses in part through regulation of protein-protein and protein-DNA interactions. The TP53 gene frequently is mutated in cancers, and in contrast to most other tumor suppressors, the mutations are mostly missense often resulting in the accumulation of mutant (MUT) protein, which may have novel or altered functions. Most MUT TP53s can be posttranslationally modified at the same residues as in WT TP53. Strikingly, however, codons for modified residues are rarely mutated in human tumors, suggesting that TP53 modifications are not essential for tumor suppression activity. Nevertheless, these modifications might alter MUT TP53 activity and contribute to a gain-of-function leading to increased metastasis and tumor progression. Furthermore, many of the signal transduction pathways that result in TP53 modifications are altered or disrupted in cancers. Understanding the signaling pathways that result in TP53 modification and the functions of these modifications in both WT TP53 and its many MUT forms may contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Thuy-Ai Nguyen
- Chromosome Stability Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | |
Collapse
|
23
|
Tsuchiya A, Kanno T, Shimizu T, Nakao S, Tanaka A, Tabata C, Nakano T, Nishizaki T. A novel PP2A enhancer induces caspase-independent apoptosis of MKN28 gastric cancer cells with high MEK activity. Cancer Lett 2014; 347:123-8. [PMID: 24508028 DOI: 10.1016/j.canlet.2014.01.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 01/17/2014] [Accepted: 01/29/2014] [Indexed: 11/27/2022]
Abstract
The newly synthesized phosphatidylinositol (PI) derivative 1,2-O-bis-[8-{2-(2-pentyl-cyclopropylmethyl)-cyclopropyl}-octanoyl]-sn-glycero-3-phosphatidyl-D-1-inositol (diDCP-LA-PI) significantly enhanced protein phosphatase 2A (PP2A) activity in the cell-free assay. This prompted to assess the antitumor effect of diDCP-LA-PI. diDCP-LA-PI attenuated phosphorylation of mitogen-activated protein kinase (MAPK) kinase (MEK) in Lu65 human lung cancer and MKN28 human gastric cancer cells with high MEK activity. diDCP-LA-PI reduced cell viability in Lu65 and MKN28 cells, but otherwise such effect was not found in 786-O human renal cancer and HUH-7 human hepatoma cells with relatively low MEK activity. For Lu65 and MKN28 cells diDCP-LA-PI increased terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells, but no significant activation of caspase-3, -8, or -9 was obtained. For MKN28 cells diDCP-LA-PI-induced reduction of MEK phosphorylation and cell viability was prevented by knocking-down PP2Ac. Taken together, these results indicate that diDCP-LA-PI induces caspase-independent apoptosis of Lu65 and MKN28 human cancer cells, for the latter cells by suppressing MEK activity through PP2A-catalyzed dephosphorylation.
Collapse
Affiliation(s)
- Ayako Tsuchiya
- Division of Bioinformation, Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan
| | - Takeshi Kanno
- Division of Bioinformation, Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan
| | - Tadashi Shimizu
- Laboratory of Chemical Biology, Advanced Medicinal Research Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe 650-8530, Japan
| | - Syuhei Nakao
- Laboratory of Chemical Biology, Advanced Medicinal Research Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe 650-8530, Japan
| | - Akito Tanaka
- Laboratory of Chemical Biology, Advanced Medicinal Research Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe 650-8530, Japan
| | - Chiharu Tabata
- Department of Thoracic Oncology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan
| | - Takashi Nakano
- Department of Thoracic Oncology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan
| | - Tomoyuki Nishizaki
- Division of Bioinformation, Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan.
| |
Collapse
|
24
|
Activation of p107 by fibroblast growth factor, which is essential for chondrocyte cell cycle exit, is mediated by the protein phosphatase 2A/B55α holoenzyme. Mol Cell Biol 2013; 33:3330-42. [PMID: 23775125 DOI: 10.1128/mcb.00082-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The phosphorylation state of pocket proteins during the cell cycle is determined at least in part by an equilibrium between inducible cyclin-dependent kinases (CDKs) and serine/threonine protein phosphatase 2A (PP2A). Two trimeric holoenzymes consisting of the core PP2A catalytic/scaffold dimer and either the B55α or PR70 regulatory subunit have been implicated in the activation of p107/p130 and pRB, respectively. While the phosphorylation state of p107 is very sensitive to forced changes of B55α levels in human cell lines, regulation of p107 in response to physiological modulation of PP2A/B55α has not been elucidated. Here we show that fibroblast growth factor 1 (FGF1), which induces maturation and cell cycle exit in chondrocytes, triggers rapid accumulation of p107-PP2A/B55α complexes coinciding with p107 dephosphorylation. Reciprocal solution-based mass spectrometric analysis identified the PP2A/B55α complex as a major component in p107 complexes, which also contain E2F/DPs, DREAM subunits, and/or cyclin/CDK complexes. Of note, p107 is one of the preferred partners of B55α, which also associates with pRB in RCS cells. FGF1-induced dephosphorylation of p107 results in its rapid accumulation in the nucleus and formation of larger complexes containing p107 and enhances its interaction with E2F4 and other p107 partners. Consistent with a key role of B55α in the rapid activation of p107 in chondrocytes, limited ectopic expression of B55α results in marked dephosphorylation of p107 while B55α knockdown results in hyperphosphorylation. More importantly, knockdown of B55α dramatically delays FGF1-induced dephosphorylation of p107 and slows down cell cycle exit. Moreover, dephosphorylation of p107 in response to FGF1 treatment results in early recruitment of p107 to the MYC promoter, an FGF1/E2F-regulated gene. Our results suggest a model in which FGF1 mediates rapid dephosphorylation and activation of p107 independently of the CDK activities that maintain p130 and pRB hyperphosphorylation for several hours after p107 dephosphorylation in maturing chondrocytes.
Collapse
|