1
|
Lin L, Ding J, Liu S, Liu C, Li Q, Gao X, Niu Y, Tong WM. Protein Phosphatase 2ACα Regulates ATR-Mediated Endogenous DNA Damage Response Against Microcephaly. Mol Neurobiol 2024:10.1007/s12035-024-04301-6. [PMID: 38976130 DOI: 10.1007/s12035-024-04301-6] [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: 11/29/2022] [Accepted: 06/11/2024] [Indexed: 07/09/2024]
Abstract
Protein phosphatase 2A (PP2A) is an abundant heterotrimeric holoenzyme in eukaryotic cells coordinating with specific kinases to regulate spatial-temporal protein dephosphorylation in various biological processes. However, the function of PP2A in cortical neurogenesis remains largely unknown. Here, we report that neuronal-specific deletion of Pp2acα in mice displayed microcephaly, with significantly smaller brains and defective learning and memory ability. Mechanistically, neuronal Pp2acα deficiency resulted in elevated endogenous DNA damage and activation of ATR/CHK1 signaling. It was further induced by the loss of direct interaction between PP2AC and ATR as well as the function of PP2AC to dephosphorylate ATR. Importantly, ATR/CHK1 signaling dysregulation altered both the expression and activity of several critical downstream factors including P53, P21, Bcl2, and Bax, which led to decreased proliferation of cortical progenitor cells and increased apoptosis in developing cortical neurons. Taken together, our results indicate an essential function of PP2ACα in endogenous DNA damage response-mediated ATR signaling during neurogenesis, and defective PP2ACα in neurons contributes to microcephaly.
Collapse
Affiliation(s)
- Lin Lin
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jing Ding
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Simeng Liu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Department of Pathology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chunying Liu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Qing Li
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Yamei Niu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Sule A, Golding SE, Ahmad SF, Watson J, Ahmed MH, Kellogg GE, Bernas T, Koebley S, Reed JC, Povirk LF, Valerie K. ATM phosphorylates PP2A subunit A resulting in nuclear export and spatiotemporal regulation of the DNA damage response. Cell Mol Life Sci 2022; 79:603. [PMID: 36434396 PMCID: PMC9700600 DOI: 10.1007/s00018-022-04550-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022]
Abstract
Ataxia telangiectasia mutated (ATM) is a serine-threonine protein kinase and important regulator of the DNA damage response (DDR). One critical ATM target is the structural subunit A (PR65-S401) of protein phosphatase 2A (PP2A), known to regulate diverse cellular processes such as mitosis and cell growth as well as dephosphorylating many proteins during the recovery from the DDR. We generated mouse embryonic fibroblasts expressing PR65-WT, -S401A (cannot be phosphorylated), and -S401D (phospho-mimetic) transgenes. Significantly, S401 mutants exhibited extensive chromosomal aberrations, impaired DNA double-strand break (DSB) repair and underwent increased mitotic catastrophe after radiation. Both S401A and the S401D cells showed impaired DSB repair (nonhomologous end joining and homologous recombination repair) and exhibited delayed DNA damage recovery, which was reflected in reduced radiation survival. Furthermore, S401D cells displayed increased ERK and AKT signaling resulting in enhanced growth rate further underscoring the multiple roles ATM-PP2A signaling plays in regulating prosurvival responses. Time-lapse video and cellular localization experiments showed that PR65 was exported to the cytoplasm after radiation by CRM1, a nuclear export protein, in line with the very rapid pleiotropic effects observed. A putative nuclear export sequence (NES) close to S401 was identified and when mutated resulted in aberrant PR65 shuttling. Our study demonstrates that the phosphorylation of a single, critical PR65 amino acid (S401) by ATM fundamentally controls the DDR, and balances DSB repair quality, cell survival and growth by spatiotemporal PR65 nuclear-cytoplasmic shuttling mediated by the nuclear export receptor CRM1.
Collapse
Affiliation(s)
- Amrita Sule
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, 23298-0058, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Sarah E Golding
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, 23298-0058, USA
| | - Syed F Ahmad
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, 23298-0058, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - James Watson
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, 23298-0058, USA
| | - Mostafa H Ahmed
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Glen E Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Tytus Bernas
- Department of Anatomy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Sean Koebley
- Department of Physics, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Jason C Reed
- Department of Physics, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Lawrence F Povirk
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Kristoffer Valerie
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, 23298-0058, USA.
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
3
|
Madduri LSV, Brandquist ND, Palanivel C, Talmon GA, Baine MJ, Zhou S, Enke CA, Johnson KR, Ouellette MM, Yan Y. p53/FBXL20 axis negatively regulates the protein stability of PR55α, a regulatory subunit of PP2A Ser/Thr phosphatase. Neoplasia 2021; 23:1192-1203. [PMID: 34731788 PMCID: PMC8570931 DOI: 10.1016/j.neo.2021.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/02/2021] [Accepted: 10/13/2021] [Indexed: 11/25/2022]
Abstract
We have previously reported an important role of PR55α, a regulatory subunit of PP2A Ser/Thr phosphatase, in the support of critical oncogenic pathways required for oncogenesis and the malignant phenotype of pancreatic cancer. The studies in this report reveal a novel mechanism by which the p53 tumor suppressor inhibits the protein-stability of PR55α via FBXL20, a p53-target gene that serves as a substrate recognition component of the SCF (Skp1_Cullin1_F-box) E3 ubiquitin ligase complex that promotes proteasomal degradation of its targeted proteins. Our studies show that inactivation of p53 by siRNA-knockdown, gene-deletion, HPV-E6-mediated degradation, or expression of the loss-of-function mutant p53R175H results in increased PR55α protein stability, which is accompanied by reduced protein expression of FBXL20 and decreased ubiquitination of PR55α. Subsequent studies demonstrate that knockdown of FBXL20 by siRNA mimics p53 deficiency, reducing PR55α ubiquitination and increasing PR55α protein stability. Functional tests indicate that ectopic p53R175H or PR55α expression results in an increase of c-Myc protein stability with concomitant dephosphorylation of c-Myc-T58, which is a PR55α substrate, whose phosphorylation otherwise promotes c-Myc degradation. A significant increase in anchorage-independent proliferation is also observed in normal human pancreatic cells expressing p53R175H or, to a greater extent, overexpressing PR55α. Consistent with the common loss of p53 function in pancreatic cancer, FBXL20 mRNA expression is significantly lower in pancreatic cancer tissues compared to pancreatic normal tissues and low FBXL20 levels correlate with poor patient survival. Collectively, these studies delineate a novel mechanism by which the p53/FBXL20 axis negatively regulates PR55α protein stability.
Collapse
Affiliation(s)
- Lepakshe S V Madduri
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA
| | - Nichole D Brandquist
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA
| | - Chitra Palanivel
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael J Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sumin Zhou
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA
| | - Charles A Enke
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA
| | - Keith R Johnson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; College of Dentistry-Oral Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michel M Ouellette
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, 986850 Nebraska Medical Center, Omaha, NE 68198-6850, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Pesch AM, Pierce LJ, Speers CW. Modulating the Radiation Response for Improved Outcomes in Breast Cancer. JCO Precis Oncol 2021; 5:PO.20.00297. [PMID: 34250414 DOI: 10.1200/po.20.00297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/12/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022] Open
Affiliation(s)
- Andrea M Pesch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Department of Pharmacology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Lori J Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Corey W Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
5
|
Papke CM, Smolen KA, Swingle MR, Cressey L, Heng RA, Toporsian M, Deng L, Hagen J, Shen Y, Chung WK, Kettenbach AN, Honkanen RE. A disorder-related variant (E420K) of a PP2A-regulatory subunit (PPP2R5D) causes constitutively active AKT-mTOR signaling and uncoordinated cell growth. J Biol Chem 2021; 296:100313. [PMID: 33482199 PMCID: PMC7952134 DOI: 10.1016/j.jbc.2021.100313] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/08/2023] Open
Abstract
Functional genomic approaches have facilitated the discovery of rare genetic disorders and improved efforts to decipher their underlying etiology. PPP2R5D-related disorder is an early childhood onset condition characterized by intellectual disability, hypotonia, autism-spectrum disorder, macrocephaly, and dysmorphic features. The disorder is caused by de novo single nucleotide changes in PPP2R5D, which generate heterozygous dominant missense variants. PPP2R5D is known to encode a B'-type (B'56δ) regulatory subunit of a PP2A-serine/threonine phosphatase. To help elucidate the molecular mechanisms altered in PPP2R5D-related disorder, we used a CRISPR-single-base editor to generate HEK-293 cells in which a single transition (c.1258G>A) was introduced into one allele, precisely recapitulating a clinically relevant E420K variant. Unbiased quantitative proteomic and phosphoproteomic analyses of endogenously expressed proteins revealed heterozygous-dominant changes in kinase/phosphatase signaling. These data combined with orthogonal validation studies revealed a previously unrecognized interaction of PPP2R5D with AKT in human cells, leading to constitutively active AKT-mTOR signaling, increased cell size, and uncoordinated cellular growth in E420K-variant cells. Rapamycin reduced cell size and dose-dependently reduced RPS6 phosphorylation in E420K-variant cells, suggesting that inhibition of mTOR1 can suppress both the observed RPS6 hyperphosphorylation and increased cell size. Together, our findings provide a deeper understanding of PPP2R5D and insight into how the E420K-variant alters signaling networks influenced by PPP2R5D. Our comprehensive approach, which combines precise genome editing, isobaric tandem mass tag labeling of peptides generated from endogenously expressed proteins, and concurrent liquid chromatography-mass spectrometry (LC-MS3), also provides a roadmap that can be used to rapidly explore the etiologies of additional genetic disorders.
Collapse
Affiliation(s)
- Cinta M Papke
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Kali A Smolen
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Mark R Swingle
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Lauren Cressey
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Richard A Heng
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Mourad Toporsian
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Liyong Deng
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Jacob Hagen
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA; Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA.
| | - Richard E Honkanen
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
6
|
Saha N, Muntean AG. Insight into the multi-faceted role of the SUV family of H3K9 methyltransferases in carcinogenesis and cancer progression. Biochim Biophys Acta Rev Cancer 2020; 1875:188498. [PMID: 33373647 DOI: 10.1016/j.bbcan.2020.188498] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022]
Abstract
Growing evidence implicates histone H3 lysine 9 methylation in tumorigenesis. The SUV family of H3K9 methyltransferases, which include G9a, GLP, SETDB1, SETDB2, SUV39H1 and SUV39H2 deposit H3K9me1/2/3 marks at euchromatic and heterochromatic regions, catalyzed by their conserved SET domain. In cancer, this family of enzymes can be deregulated by genomic alterations and transcriptional mis-expression leading to alteration of transcriptional programs. In solid and hematological malignancies, studies have uncovered pro-oncogenic roles for several H3K9 methyltransferases and accordingly, small molecule inhibitors are being tested as potential therapies. However, emerging evidence demonstrate onco-suppressive roles for these enzymes in cancer development as well. Here, we review the role H3K9 methyltransferases play in tumorigenesis focusing on gene targets and biological pathways affected due to misregulation of these enzymes. We also discuss molecular mechanisms regulating H3K9 methyltransferases and their influence on cancer. Finally, we describe the impact of H3K9 methylation on therapy induced resistance in carcinoma. Converging evidence point to multi-faceted roles for H3K9 methyltransferases in development and cancer that encourages a deeper understanding of these enzymes to inform novel therapy.
Collapse
Affiliation(s)
- Nirmalya Saha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| | - Andrew G Muntean
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America.
| |
Collapse
|
7
|
Li F, Kozono D, Deraska P, Branigan T, Dunn C, Zheng XF, Parmar K, Nguyen H, DeCaprio J, Shapiro GI, Chowdhury D, D'Andrea AD. CHK1 Inhibitor Blocks Phosphorylation of FAM122A and Promotes Replication Stress. Mol Cell 2020; 80:410-422.e6. [PMID: 33108758 DOI: 10.1016/j.molcel.2020.10.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/14/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022]
Abstract
While effective anti-cancer drugs targeting the CHK1 kinase are advancing in the clinic, drug resistance is rapidly emerging. Here, we demonstrate that CRISPR-mediated knockout of the little-known gene FAM122A/PABIR1 confers cellular resistance to CHK1 inhibitors (CHK1is) and cross-resistance to ATR inhibitors. Knockout of FAM122A results in activation of PP2A-B55α, a phosphatase that dephosphorylates the WEE1 protein and rescues WEE1 from ubiquitin-mediated degradation. The resulting increase in WEE1 protein expression reduces replication stress, activates the G2/M checkpoint, and confers cellular resistance to CHK1is. Interestingly, in tumor cells with oncogene-driven replication stress, CHK1 can directly phosphorylate FAM122A, leading to activation of the PP2A-B55α phosphatase and increased WEE1 expression. A combination of a CHK1i plus a WEE1 inhibitor can overcome CHK1i resistance of these tumor cells, thereby enhancing anti-cancer activity. The FAM122A expression level in a tumor cell can serve as a useful biomarker for predicting CHK1i sensitivity or resistance.
Collapse
Affiliation(s)
- Feng Li
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Peter Deraska
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Timothy Branigan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 01115
| | - Connor Dunn
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xiao-Feng Zheng
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kalindi Parmar
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Huy Nguyen
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - James DeCaprio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 01115
| | - Geoffrey I Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 01115; Early Drug Development Center, Dana-Farber Cancer Institute, Boston, MA 02215
| | - Dipanjan Chowdhury
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
8
|
Lei X, Ma N, Du L, Liang Y, Zhang P, Han Y, Qu B. PP2A and tumor radiotherapy. Hereditas 2020; 157:36. [PMID: 32847617 PMCID: PMC7450598 DOI: 10.1186/s41065-020-00149-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that serves as a key regulator of cellular physiology in the context of apoptosis, mitosis, and DNA damage responses. Canonically, PP2A functions as a tumor suppressor gene. However, recent evidence suggests that inhibiting PP2A activity in tumor cells may represent a viable approach to enhancing tumor sensitivity to chemoradiotherapy as such inhibition can cause cells to enter a disordered mitotic state that renders them more susceptible to cell death. Indeed, there is evidence that inhibiting PP2A can slow tumor growth following radiotherapy in a range of cancer types including ovarian cancer, liver cancer, malignant glioma, pancreatic cancer, and nasopharyngeal carcinoma. In the present review, we discuss current understanding of the role of PP2A in tumor radiotherapy and the potential mechanisms whereby it may influence this process.
Collapse
Affiliation(s)
- Xiao Lei
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China
| | - Na Ma
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China
| | - Lehui Du
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China
| | - Yanjie Liang
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China
| | - Pei Zhang
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China
| | - Yanan Han
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China
| | - Baolin Qu
- The First Medical Center of Chinese PLA General Hospital, Department of Radiation Oncology, Beijing, P. R. China.
| |
Collapse
|
9
|
Cell Cycle and DNA Repair Regulation in the Damage Response: Protein Phosphatases Take Over the Reins. Int J Mol Sci 2020; 21:ijms21020446. [PMID: 31936707 PMCID: PMC7014277 DOI: 10.3390/ijms21020446] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/29/2019] [Accepted: 01/02/2020] [Indexed: 12/14/2022] Open
Abstract
Cells are constantly suffering genotoxic stresses that affect the integrity of our genetic material. Genotoxic insults must be repaired to avoid the loss or inappropriate transmission of the genetic information, a situation that could lead to the appearance of developmental abnormalities and tumorigenesis. To combat this threat, eukaryotic cells have evolved a set of sophisticated molecular mechanisms that are collectively known as the DNA damage response (DDR). This surveillance system controls several aspects of the cellular response, including the detection of lesions, a temporary cell cycle arrest, and the repair of the broken DNA. While the regulation of the DDR by numerous kinases has been well documented over the last decade, the complex roles of protein dephosphorylation have only recently begun to be investigated. Here, we review recent progress in the characterization of DDR-related protein phosphatases during the response to a DNA lesion, focusing mainly on their ability to modulate the DNA damage checkpoint and the repair of the damaged DNA. We also discuss their protein composition and structure, target specificity, and biochemical regulation along the different stages encompassed in the DDR. The compilation of this information will allow us to better comprehend the physiological significance of protein dephosphorylation in the maintenance of genome integrity and cell viability in response to genotoxic stress.
Collapse
|
10
|
Hein AL, Brandquist ND, Ouellette CY, Seshacharyulu P, Enke CA, Ouellette MM, Batra SK, Yan Y. PR55α regulatory subunit of PP2A inhibits the MOB1/LATS cascade and activates YAP in pancreatic cancer cells. Oncogenesis 2019; 8:63. [PMID: 31659153 PMCID: PMC6817822 DOI: 10.1038/s41389-019-0172-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022] Open
Abstract
PP2A holoenzyme complexes are responsible for the majority of Ser/Thr phosphatase activities in human cells. Each PP2A consists of a catalytic subunit (C), a scaffold subunit (A), and a regulatory subunit (B). While the A and C subunits each exists only in two highly conserved isoforms, a large number of B subunits share no homology, which determines PP2A substrate specificity and cellular localization. It is anticipated that different PP2A holoenzymes play distinct roles in cellular signaling networks, whereas PP2A has only generally been defined as a putative tumor suppressor, which is mostly based on the loss-of-function studies using pharmacological or biological inhibitors for the highly conserved A or C subunit of PP2A. Recent studies of specific pathways indicate that some PP2A complexes also possess tumor-promoting functions. We have previously reported an essential role of PR55α, a PP2A regulatory subunit, in the support of oncogenic phenotypes, including in vivo tumorigenicity/metastasis of pancreatic cancer cells. In this report, we have elucidated a novel role of PR55α-regulated PP2A in the activation of YAP oncoprotein, whose function is required for anchorage-independent growth during oncogenesis of solid tumors. Our data show two lines of YAP regulation by PR55α: (1) PR55α inhibits the MOB1-triggered autoactivation of LATS1/2 kinases, the core member of the Hippo pathway that inhibits YAP by inducing its proteasomal degradation and cytoplasmic retention and (2) PR55α directly interacts with and regulates YAP itself. Accordingly, PR55α is essential for YAP-promoted gene transcriptions, as well as for anchorage-independent growth, in which YAP plays a key role. In summary, current findings demonstrate a novel YAP activation mechanism based on the PR55α-regulated PP2A phosphatase.
Collapse
Affiliation(s)
- Ashley L Hein
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nichole D Brandquist
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Caroline Y Ouellette
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Charles A Enke
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michel M Ouellette
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
11
|
Ramos F, Villoria MT, Alonso-Rodríguez E, Clemente-Blanco A. Role of protein phosphatases PP1, PP2A, PP4 and Cdc14 in the DNA damage response. Cell Stress 2019; 3:70-85. [PMID: 31225502 PMCID: PMC6551743 DOI: 10.15698/cst2019.03.178] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Maintenance of genome integrity is fundamental for cellular physiology. Our hereditary information encoded in the DNA is intrinsically susceptible to suffer variations, mostly due to the constant presence of endogenous and environmental genotoxic stresses. Genomic insults must be repaired to avoid loss or inappropriate transmission of the genetic information, a situation that could lead to the appearance of developmental anomalies and tumorigenesis. To safeguard our genome, cells have evolved a series of mechanisms collectively known as the DNA damage response (DDR). This surveillance system regulates multiple features of the cellular response, including the detection of the lesion, a transient cell cycle arrest and the restoration of the broken DNA molecule. While the role of multiple kinases in the DDR has been well documented over the last years, the intricate roles of protein dephosphorylation have only recently begun to be addressed. In this review, we have compiled recent information about the function of protein phosphatases PP1, PP2A, PP4 and Cdc14 in the DDR, focusing mainly on their capacity to regulate the DNA damage checkpoint and the repair mechanism encompassed in the restoration of a DNA lesion.
Collapse
Affiliation(s)
- Facundo Ramos
- Cell Cycle and Genome Stability Group. Institute of Functional Biology and Genomics (IBFG). Spanish National Research Council (CSIC), University of Salamanca (USAL), C/Zacarías González 2, Salamanca 37007, SPAIN
| | - María Teresa Villoria
- Cell Cycle and Genome Stability Group. Institute of Functional Biology and Genomics (IBFG). Spanish National Research Council (CSIC), University of Salamanca (USAL), C/Zacarías González 2, Salamanca 37007, SPAIN
| | - Esmeralda Alonso-Rodríguez
- Cell Cycle and Genome Stability Group. Institute of Functional Biology and Genomics (IBFG). Spanish National Research Council (CSIC), University of Salamanca (USAL), C/Zacarías González 2, Salamanca 37007, SPAIN
| | - Andrés Clemente-Blanco
- Cell Cycle and Genome Stability Group. Institute of Functional Biology and Genomics (IBFG). Spanish National Research Council (CSIC), University of Salamanca (USAL), C/Zacarías González 2, Salamanca 37007, SPAIN
| |
Collapse
|
12
|
Mazhar S, Taylor SE, Sangodkar J, Narla G. Targeting PP2A in cancer: Combination therapies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:51-63. [PMID: 30401535 DOI: 10.1016/j.bbamcr.2018.08.020] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/21/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022]
Abstract
The serine/threonine phosphatase PP2A regulates a vast portion of the phosphoproteome including pathways involved in apoptosis, proliferation and DNA damage response and PP2A inactivation is a vital step in malignant transformation. Many groups have explored the therapeutic venue of combining PP2A reactivation with kinase inhibition to counteract the very changes in tumor suppressors and oncogenes that lead to cancer development. Conversely, inhibition of PP2A to complement chemotherapy and radiation-induced cancer cell death is also an area of active investigation. Here we review the studies that utilize PP2A targeted agents as combination therapy in cancer. A potential role for PP2A in tumor immunity is also highlighted.
Collapse
Affiliation(s)
- Sahar Mazhar
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah E Taylor
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Jaya Sangodkar
- Division of Genetic Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Goutham Narla
- Division of Genetic Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Göder A, Emmerich C, Nikolova T, Kiweler N, Schreiber M, Kühl T, Imhof D, Christmann M, Heinzel T, Schneider G, Krämer OH. HDAC1 and HDAC2 integrate checkpoint kinase phosphorylation and cell fate through the phosphatase-2A subunit PR130. Nat Commun 2018; 9:764. [PMID: 29472538 PMCID: PMC5823910 DOI: 10.1038/s41467-018-03096-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 01/19/2018] [Indexed: 12/18/2022] Open
Abstract
Checkpoint kinases sense replicative stress to prevent DNA damage. Here we show that the histone deacetylases HDAC1/HDAC2 sustain the phosphorylation of the checkpoint kinases ATM, CHK1 and CHK2, activity of the cell cycle gatekeeper kinases WEE1 and CDK1, and induction of the tumour suppressor p53 in response to stalled DNA replication. Consequently, HDAC inhibition upon replicative stress promotes mitotic catastrophe. Mechanistically, HDAC1 and HDAC2 suppress the expression of PPP2R3A/PR130, a regulatory subunit of the trimeric serine/threonine phosphatase 2 (PP2A). Genetic elimination of PR130 reveals that PR130 promotes dephosphorylation of ATM by PP2A. Moreover, the ablation of PR130 slows G1/S phase transition and increases the levels of phosphorylated CHK1, replication protein A foci and DNA damage upon replicative stress. Accordingly, stressed PR130 null cells are very susceptible to HDAC inhibition, which abrogates the S phase checkpoint, induces apoptosis and reduces the homologous recombination protein RAD51. Thus, PR130 controls cell fate decisions upon replicative stress. Checkpoint kinases control cell cycle progression via the regulation of many key regulators. Here the authors demonstrate how HDAC1 and HDAC2 modulate checkpoint kinase signalling via the suppression of PR130, a regulatory subunit of the trimeric serine/threonine phosphatase 2.
Collapse
Affiliation(s)
- Anja Göder
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Claudia Emmerich
- University of Jena, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Hans-Knöll-Strasse 2, 07745, Jena, Germany
| | - Teodora Nikolova
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Nicole Kiweler
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Maria Schreiber
- University of Jena, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Hans-Knöll-Strasse 2, 07745, Jena, Germany
| | - Toni Kühl
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Markus Christmann
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | - Thorsten Heinzel
- University of Jena, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Hans-Knöll-Strasse 2, 07745, Jena, Germany
| | - Günter Schneider
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Ismaningerstrasse 22, 81675, Munich, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany.
| |
Collapse
|
14
|
Baumgartner U, Berger F, Hashemi Gheinani A, Burgener SS, Monastyrskaya K, Vassella E. miR-19b enhances proliferation and apoptosis resistance via the EGFR signaling pathway by targeting PP2A and BIM in non-small cell lung cancer. Mol Cancer 2018; 17:44. [PMID: 29455644 PMCID: PMC5817797 DOI: 10.1186/s12943-018-0781-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/01/2018] [Indexed: 12/19/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) mutations enable constitutive active downstream signaling of PI3K/AKT, KRAS/ERK and JAK/STAT pathways, and promote tumor progression by inducing uncontrolled proliferation, evasion of apoptosis and migration of non-small cell lung cancer (NSCLC). In addition, such EGFR mutations increase the susceptibility of patients with NSCLC to tyrosine kinase inhibitor (TKI) therapy, but treated patients will invariably relapse with resistant disease. A global understanding of underlying molecular mechanisms of EGFR signaling may improve the management of NSCLC patients. Methods microarray analysis was performed to identify PI3K/AKT-regulated miRNAs. Phosphoproteomic analysis and cell based assays were performed using NSCLC cell lines lentivirally transduced with anti-miR or miR overexpressing constructs. Results Here, we show that 17 miRNAs including members of the miR-17~ 92 cluster are dysregulated following PI3K/AKT inhibition of EGFR mutant NSCLC cells. Bioinformatics analysis revealed that dysregulated miRNAs act in a concerted manner to enhance the activity of the EGFR signaling pathway. These findings were closely mirrored by attenuation of miR-17~ 92 family member miR-19b in NSCLC cell lines which resulted in reduced phosphorylation of ERK, AKT and STAT and effector proteins in EGFR mutant NSCLC cells. Consistent with this finding, cell cycle progression, clonogenic growth and migration were reduced and apoptosis was enhanced. Co-treatment of NSCLC cells with the tyrosine kinase inhibitor (TKI) gefitinib and anti-miR-19b construct reduced migration and clonogenic growth in a synergistic manner suggesting that EGFR and miR-19b act together to control oncogenic processes. Serine/threonine phosphatase PP2A subunit PPP2R5E and BCL2L11 encoding BIM were identified as major targets of miR-19b by target validation assays. Consistent with this finding, PP2A activity was strongly enhanced in NSCLC transduced with anti-miR-19b construct, but not in cells co-transduced with anti-miR-19b and shPPP2R5E, suggesting that PPP2R5E is a major constituent of the PP2A complex. Accordingly, enhanced proliferation by miR-19b was due to targeting PPP2R5E. In contrast, apoptosis resistance was mainly due to targeting BCL2L11. Conclusion Our results provide insight into the importance of targeting PPP2R5E and BCL2L11 by miR-19b in oncogenic processes of NSCLC. Attenuation of miR-19b expression could potentially be exploited in adjuvant therapy of EGFR mutant NSCLC. Electronic supplementary material The online version of this article (10.1186/s12943-018-0781-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ulrich Baumgartner
- Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Fabienne Berger
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Sabrina Sofia Burgener
- Institute for Virology and Immunology, Vetsuisse Faculty, University of Bern, Mittelhäusern, Bern, Switzerland
| | | | - Erik Vassella
- Institute of Pathology, University of Bern, Bern, Switzerland. .,Institut für Pathologie, University of Bern, Murtenstrasse 31, CH-3008, Bern, Switzerland.
| |
Collapse
|
15
|
Sales Gil R, de Castro IJ, Berihun J, Vagnarelli P. Protein phosphatases at the nuclear envelope. Biochem Soc Trans 2018; 46:173-182. [PMID: 29432143 PMCID: PMC5818667 DOI: 10.1042/bst20170139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 12/14/2022]
Abstract
The nuclear envelope (NE) is a unique topological structure formed by lipid membranes (Inner and Outer Membrane: IM and OM) interrupted by open channels (Nuclear Pore complexes). Besides its well-established structural role in providing a physical separation between the genome and the cytoplasm and regulating the exchanges between the two cellular compartments, it has become quite evident in recent years that the NE also represents a hub for localized signal transduction. Mechanical, stress, or mitogen signals reach the nucleus and trigger the activation of several pathways, many effectors of which are processed at the NE. Therefore, the concept of the NE acting just as a barrier needs to be expanded to embrace all the dynamic processes that are indeed associated with it. In this context, dynamic protein association and turnover coupled to reversible post-translational modifications of NE components can provide important clues on how this integrated cellular machinery functions as a whole. Reversible protein phosphorylation is the most used mechanism to control protein dynamics and association in cells. Keys to the reversibility of the system are protein phosphatases and the regulation of their activity in space and time. As the NE is clearly becoming an interesting compartment for the control and transduction of several signalling pathways, in this review we will focus on the role of Protein Phosphatases at the NE since the significance of this class of proteins in this context has been little explored.
Collapse
Affiliation(s)
- Raquel Sales Gil
- College of Health and Life Science, Research Institute for Environment Health and Society, Brunel University London, London UB8 3PH, U.K
| | - Ines J de Castro
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg and German Center for Infection Research (DZIF), Heidelberg 69120, Germany
| | - Jerusalem Berihun
- College of Health and Life Science, Research Institute for Environment Health and Society, Brunel University London, London UB8 3PH, U.K
| | - Paola Vagnarelli
- College of Health and Life Science, Research Institute for Environment Health and Society, Brunel University London, London UB8 3PH, U.K.
| |
Collapse
|
16
|
LB-100, a novel Protein Phosphatase 2A (PP2A) inhibitor, sensitizes malignant meningioma cells to the therapeutic effects of radiation. Cancer Lett 2017; 415:217-226. [PMID: 29199006 DOI: 10.1016/j.canlet.2017.11.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Atypical and anaplastic meningiomas (AAM) represent 20% of all meningiomas. They are associated with poor outcomes due to their tendency to recur. While surgery and radiation (RT) are first line therapy, no effective systemic medical treatment has been identified. Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase involved in cell cycle regulation and DNA repair. Here, we examined radiosensitizing effects of LB-100, a novel inhibitor of PP2A against AAM as a novel treatment strategy. Three human-derived immortalized meningioma cell lines, IOMM-LEE, GAR, and CH-157, were used to investigate the radio-sensitizing potential of LB-100 in AAM. Survival fraction by clonogenic assay, immunofluorescence, cell cycle analysis and protein expression were evaluated in vitro. The antitumor effects of combining LB-100 with RT were verified in vivo by using intracranial orthotopic xenograft mouse model. Pharmacologic PP2A inhibition with LB-100 prior to RT enhanced the radiosensitivity of meningioma cells and reduced survival fraction in clonogenic assays. LB-100 increased DNA double-strand breakage (measured by γ-H2AX), mitotic catastrophe cell death, and G2/M cell cycle arrest in irradiated meningioma cells. Also, LB-100 decreased activation of STAT3 and expression of its downstream proteins. In vivo, LB-100 and RT combined treatment prolonged the survival of mice with xenografts compared to RT alone. Taken together, these results provide convincing preclinical data to support the use of LB-100 as a radiosensitizing agent for treatment of malignant meningioma. Its potential for clinical application deserves further investigation.
Collapse
|
17
|
Phosphatases and solid tumors: focus on glioblastoma initiation, progression and recurrences. Biochem J 2017; 474:2903-2924. [PMID: 28801478 DOI: 10.1042/bcj20170112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022]
Abstract
Phosphatases and cancer have been related for many years now, as these enzymes regulate key cellular functions, including cell survival, migration, differentiation and proliferation. Dysfunctions or mutations affecting these enzymes have been demonstrated to be key factors for oncogenesis. The aim of this review is to shed light on the role of four different phosphatases (PTEN, PP2A, CDC25 and DUSP1) in five different solid tumors (breast cancer, lung cancer, pancreatic cancer, prostate cancer and ovarian cancer), in order to better understand the most frequent and aggressive primary cancer of the central nervous system, glioblastoma.
Collapse
|
18
|
Sankhe S, Manousakidi S, Antigny F, Arthur Ataam J, Bentebbal S, Ruchon Y, Lecerf F, Sabourin J, Price L, Fadel E, Dorfmüller P, Eddahibi S, Humbert M, Perros F, Capuano V. T-type Ca 2+ channels elicit pro-proliferative and anti-apoptotic responses through impaired PP2A/Akt1 signaling in PASMCs from patients with pulmonary arterial hypertension. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1631-1641. [PMID: 28655554 DOI: 10.1016/j.bbamcr.2017.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
Abstract
Idiopathic pulmonary arterial hypertension (iPAH) is characterized by obstructive hyperproliferation and apoptosis resistance of distal pulmonary artery smooth muscle cells (PASMCs). T-type Ca2+ channel blockers have been shown to reduce experimental pulmonary hypertension, although the impact of T-type channel inhibition remains unexplored in PASMCs from iPAH patients. Here we show that T-type channels Cav3.1 and Cav3.2 are present in the lung and PASMCs from iPAH patients and control subjects. The blockade of T-type channels by the specific blocker, TTA-A2, prevents cell cycle progression and PASMCs growth. In iPAH cells, T-type channel signaling fails to activate phosphatase PP2A, leading to an increase in ERK1/2, P38 activation. Moreover, T-type channel signaling is redirected towards the activation of the kinase Akt1, leading to increased expression of the anti-apoptotic protein survivin, and a decrease in the pro-apoptotic mediator FoxO3A. Finally, in iPAH cells, Akt1 is no longer able to regulate caspase 9 activation, whereas T-type channel overexpression reverses PP2A defect in iPAH cells but reinforces the deleterious effects of Akt1 activation. Altogether, these data highlight T-type channel signaling as a strong trigger of the pathological phenotype of PASMCs from iPAH patients (hyper-proliferation/cells survival and apoptosis resistance), suggesting that both T-type channels and PP2A may be promising therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Safietou Sankhe
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Sevasti Manousakidi
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Fabrice Antigny
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jennifer Arthur Ataam
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Sana Bentebbal
- PhyMedExp, Univ. Montpellier, Inserm U1046, cNRS UMR9214.34295 MINSERM U1046, Montpellier, France
| | - Yann Ruchon
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Florence Lecerf
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jessica Sabourin
- INSERM UMR-S1180, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Laura Price
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Elie Fadel
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Peter Dorfmüller
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Saadia Eddahibi
- PhyMedExp, Univ. Montpellier, Inserm U1046, cNRS UMR9214.34295 MINSERM U1046, Montpellier, France
| | - Marc Humbert
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France; AP-HP, Service de pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Frédéric Perros
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Véronique Capuano
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
19
|
de Gooijer MC, van den Top A, Bockaj I, Beijnen JH, Würdinger T, van Tellingen O. The G2 checkpoint-a node-based molecular switch. FEBS Open Bio 2017; 7:439-455. [PMID: 28396830 PMCID: PMC5377395 DOI: 10.1002/2211-5463.12206] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/09/2017] [Accepted: 01/18/2017] [Indexed: 12/20/2022] Open
Abstract
Tight regulation of the eukaryotic cell cycle is paramount to ensure genomic integrity throughout life. Cell cycle checkpoints are present in each phase of the cell cycle and prevent cell cycle progression when genomic integrity is compromised. The G2 checkpoint is an intricate signaling network that regulates the progression of G2 to mitosis (M). We propose here a node-based model of G2 checkpoint regulation, in which the action of the central CDK1-cyclin B1 node is determined by the concerted but opposing activities of the Wee1 and cell division control protein 25C (CDC25C) nodes. Phosphorylation of both Wee1 and CDC25C at specific sites determines their subcellular localization, driving them either toward activity within the nucleus or to the cytoplasm and subsequent ubiquitin-mediated proteasomal degradation. In turn, this subcellular balance of the Wee1 and CDC25C nodes is directed by the action of the PLK1 and CHK1 nodes via what we have termed the 'nuclear and cytoplasmic decision states' of Wee1 and CDC25C. The proposed node-based model provides an intelligible structure of the complex interactions that govern the decision to delay or continue G2/M progression. The model may also aid in predicting the effects of agents that target these G2 checkpoint nodes.
Collapse
Affiliation(s)
- Mark C. de Gooijer
- Division of Pharmacology/Mouse Cancer ClinicThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Arnout van den Top
- Division of Pharmacology/Mouse Cancer ClinicThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Irena Bockaj
- Division of Pharmacology/Mouse Cancer ClinicThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jos H. Beijnen
- Department of Pharmacy and PharmacologyThe Netherlands Cancer Institute/Slotervaart HospitalAmsterdamThe Netherlands
- Division of Drug ToxicologyFaculty of PharmacyUtrecht UniversityThe Netherlands
- Division of Biomedical AnalysisFaculty of ScienceUtrecht UniversityThe Netherlands
| | - Thomas Würdinger
- Neuro‐oncology Research GroupDepartments of Neurosurgery and Pediatric Oncology/HematologyCancer Center AmsterdamVU University Medical CenterThe Netherlands
- Molecular Neurogenetics UnitDepartments of Neurology and RadiologyMassachusetts General HospitalBostonMAUSA
- Neuroscience ProgramHarvard Medical SchoolBostonMAUSA
| | - Olaf van Tellingen
- Division of Pharmacology/Mouse Cancer ClinicThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
20
|
Merigliano C, Marzio A, Renda F, Somma MP, Gatti M, Vernì F. A Role for the Twins Protein Phosphatase (PP2A-B55) in the Maintenance of Drosophila Genome Integrity. Genetics 2017; 205:1151-1167. [PMID: 28040742 PMCID: PMC5340330 DOI: 10.1534/genetics.116.192781] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/21/2016] [Indexed: 01/14/2023] Open
Abstract
The protein phosphatase 2A (PP2A) is a conserved heterotrimeric enzyme that regulates several cellular processes including the DNA damage response and mitosis. Consistent with these functions, PP2A is mutated in many types of cancer and acts as a tumor suppressor. In mammalian cells, PP2A inhibition results in DNA double strand breaks (DSBs) and chromosome aberrations (CABs). However, the mechanisms through which PP2A prevents DNA damage are still unclear. Here, we focus on the role of the Drosophila twins (tws) gene in the maintenance of chromosome integrity; tws encodes the B regulatory subunit (B/B55) of PP2A. Mutations in tws cause high frequencies of CABs (0.5 CABs/cell) in Drosophila larval brain cells and lead to an abnormal persistence of γ-H2Av repair foci. However, mutations that disrupt the PP4 phosphatase activity impair foci dissolution but do not cause CABs, suggesting that a delayed foci regression is not clastogenic. We also show that Tws is required for activation of the G2/M DNA damage checkpoint while PP4 is required for checkpoint recovery, a result that points to a conserved function of these phosphatases from flies to humans. Mutations in the ATM-coding gene tefu are strictly epistatic to tws mutations for the CAB phenotype, suggesting that failure to dephosphorylate an ATM substrate(s) impairs DNA DSBs repair. In addition, mutations in the Ku70 gene, which do not cause CABs, completely suppress CAB formation in tws Ku70 double mutants. These results suggest the hypothesis that an improperly phosphorylated Ku70 protein can lead to DNA damage and CABs.
Collapse
Affiliation(s)
- Chiara Merigliano
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| | - Antonio Marzio
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| | - Fioranna Renda
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| | - Maria Patrizia Somma
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Sapienza, Università di Roma, 00185, Italy
| | - Maurizio Gatti
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Sapienza, Università di Roma, 00185, Italy
| | - Fiammetta Vernì
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| |
Collapse
|
21
|
Ferrari S, Gentili C. Maintaining Genome Stability in Defiance of Mitotic DNA Damage. Front Genet 2016; 7:128. [PMID: 27493659 PMCID: PMC4954828 DOI: 10.3389/fgene.2016.00128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/06/2016] [Indexed: 01/08/2023] Open
Abstract
The implementation of decisions affecting cell viability and proliferation is based on prompt detection of the issue to be addressed, formulation and transmission of a correct set of instructions and fidelity in the execution of orders. While the first and the last are purely mechanical processes relying on the faithful functioning of single proteins or macromolecular complexes (sensors and effectors), information is the real cue, with signal amplitude, duration, and frequency ultimately determining the type of response. The cellular response to DNA damage is no exception to the rule. In this review article we focus on DNA damage responses in G2 and Mitosis. First, we set the stage describing mitosis and the machineries in charge of assembling the apparatus responsible for chromosome alignment and segregation as well as the inputs that control its function (checkpoints). Next, we examine the type of issues that a cell approaching mitosis might face, presenting the impact of post-translational modifications (PTMs) on the correct and timely functioning of pathways correcting errors or damage before chromosome segregation. We conclude this essay with a perspective on the current status of mitotic signaling pathway inhibitors and their potential use in cancer therapy.
Collapse
Affiliation(s)
- Stefano Ferrari
- Institute of Molecular Cancer Research, University of Zurich Zurich, Switzerland
| | - Christian Gentili
- Institute of Molecular Cancer Research, University of Zurich Zurich, Switzerland
| |
Collapse
|
22
|
Hein AL, Seshacharyulu P, Rachagani S, Sheinin YM, Ouellette MM, Ponnusamy MP, Mumby MC, Batra SK, Yan Y. PR55α Subunit of Protein Phosphatase 2A Supports the Tumorigenic and Metastatic Potential of Pancreatic Cancer Cells by Sustaining Hyperactive Oncogenic Signaling. Cancer Res 2016; 76:2243-2253. [PMID: 26893480 DOI: 10.1158/0008-5472.can-15-2119] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/10/2016] [Indexed: 12/16/2022]
Abstract
The protein phosphatase 2 (PP2A) holoenzyme consists of a catalytic subunit, a scaffold subunit, and a regulatory subunit. Based on loss-of-function analysis using PP2A catalytic inhibitors or inhibition via tumor viral antigens, limited studies suggest that PP2A is a putative tumor suppressor. However, PP2A has also been shown to facilitate the activation of oncogenic signaling pathways when associated with specific regulatory subunits. In this study, we investigated the possible oncogenic role of PP2A in pancreatic cancer. We found a striking increase in the expression of PR55α (PPP2R2A), a PP2A regulatory subunit, in pancreatic cancer cells compared with normal pancreatic epithelial cells. Consistently, PR55α expression was markedly elevated in pancreatic ductal adenocarcinoma tissues compared with adjacent normal pancreatic tissues (P < 0.0001) and correlated with poor survival of pancreatic cancer patients (P < 0.0003). RNAi-mediated depletion of PR55α in pancreatic cancer cell lines resulted in diminished phosphorylation of both AKT and ERK1/2 (MAPK3/1) and decreased protein levels of β-catenin (CTNNB1). Accordingly, pancreatic cancer cells with reduced PR55α expression exhibited significantly impaired properties of transformation, including attenuated cell growth, clonogenicity, mobility, and anchorage-independent growth. Moreover, orthotopic implantation of PR55α-depleted pancreatic cancer cells into nude mice resulted in markedly reduced tumorigenicity (P < 0.001) and distant metastases. Together, these results suggest that PR55α promotes pancreatic cancer development by sustaining hyperactivity of multiple oncogenic signaling pathways, including AKT, ERK, and Wnt. These studies also provide a basis for exploring PR55α as a diagnostic or therapeutic target in pancreatic cancer. Cancer Res; 76(8); 2243-53. ©2016 AACR.
Collapse
Affiliation(s)
- Ashley L Hein
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Yuri M Sheinin
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michel M Ouellette
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Marc C Mumby
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
23
|
Ziegler V, Albers A, Fritz G. Lovastatin protects keratinocytes from DNA damage-related pro-apoptotic stress responses stimulated by anticancer therapeutics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1082-92. [PMID: 26876155 DOI: 10.1016/j.bbamcr.2016.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/29/2016] [Accepted: 02/10/2016] [Indexed: 01/14/2023]
Abstract
BACKGROUND Oral mucositis (OM) is a relevant adverse effect of anticancer therapy involving ionizing radiation (IR) and doxorubicin (Doxo). Because DNA damage of keratinocytes is causative for the pathogenesis of OM, we aim to identify pharmacological measures for geno- and cytoprotection of keratinocytes. METHODS We investigated the influence of the lipid-lowering drug lovastatin on cell death, proliferation and DNA damage response (DDR) mechanisms of human keratinocytes following treatment with IR and Doxo. RESULTS Lovastatin protected keratinocytes from the cytotoxic and genotoxic effects of IR and Doxo as shown by a diminished induction of apoptosis as well as a reduced formation and slightly improved repair of DNA damage following Doxo and IR treatment, respectively. Lovastatin selectively blocked the activation of Chk1 and ATR kinases following treatment with IR, Doxo and the ribonucleotide reductase inhibitor hydroxyurea, indicating that the statin antagonizes ATR/Chk1-regulated replicative stress responses. Part of the cytoprotective activity of lovastatin seems to rest on a delayed entry of lovastatin treated cells into S-phase. Yet, because the statin also protected non-proliferating keratinocytes from IR- and Doxo-induced cytotoxicity, cell cycle independent protective mechanisms are involved, too. CONCLUSIONS Lovastatin attenuates pro-toxic DNA damage-related responses of keratinocytes stimulated by OM-inducing anticancer therapeutics. The data encourage forthcoming in vivo and clinical studies addressing the usefulness of statins in the prevention of OM.
Collapse
Affiliation(s)
- Verena Ziegler
- Institute of Toxicology, Medical Faculty of the Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Anne Albers
- Institute of Toxicology, Medical Faculty of the Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty of the Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| |
Collapse
|
24
|
The Adenovirus E4orf4 Protein Provides a Novel Mechanism for Inhibition of the DNA Damage Response. PLoS Pathog 2016; 12:e1005420. [PMID: 26867009 PMCID: PMC4750969 DOI: 10.1371/journal.ppat.1005420] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/06/2016] [Indexed: 12/20/2022] Open
Abstract
The DNA damage response (DDR) is a conglomerate of pathways designed to detect DNA damage and signal its presence to cell cycle checkpoints and to the repair machinery, allowing the cell to pause and mend the damage, or if the damage is too severe, to trigger apoptosis or senescence. Various DDR branches are regulated by kinases of the phosphatidylinositol 3-kinase-like protein kinase family, including ataxia-telangiectasia mutated (ATM) and ATM- and Rad3-related (ATR). Replication intermediates and linear double-stranded genomes of DNA viruses are perceived by the cell as DNA damage and activate the DDR. If allowed to operate, the DDR will stimulate ligation of viral genomes and will inhibit virus replication. To prevent this outcome, many DNA viruses evolved ways to limit the DDR. As part of its attack on the DDR, adenovirus utilizes various viral proteins to cause degradation of DDR proteins and to sequester the MRN damage sensor outside virus replication centers. Here we show that adenovirus evolved yet another novel mechanism to inhibit the DDR. The E4orf4 protein, together with its cellular partner PP2A, reduces phosphorylation of ATM and ATR substrates in virus-infected cells and in cells treated with DNA damaging drugs, and causes accumulation of damaged DNA in the drug-treated cells. ATM and ATR are not mutually required for inhibition of their signaling pathways by E4orf4. ATM and ATR deficiency as well as E4orf4 expression enhance infection efficiency. Furthermore, E4orf4, previously reported to induce cancer-specific cell death when expressed alone, sensitizes cells to killing by sub-lethal concentrations of DNA damaging drugs, likely because it inhibits DNA damage repair. These findings provide one explanation for the cancer-specificity of E4orf4-induced cell death as many cancers have DDR deficiencies leading to increased reliance on the remaining intact DDR pathways and to enhanced susceptibility to DDR inhibitors such as E4orf4. Thus DDR inhibition by E4orf4 contributes both to the efficiency of adenovirus replication and to the ability of E4orf4 to kill cancer cells.
Collapse
|
25
|
Hasvold G, Lund-Andersen C, Lando M, Patzke S, Hauge S, Suo Z, Lyng H, Syljuåsen RG. Hypoxia-induced alterations of G2 checkpoint regulators. Mol Oncol 2016; 10:764-73. [PMID: 26791779 DOI: 10.1016/j.molonc.2015.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 12/23/2015] [Accepted: 12/23/2015] [Indexed: 02/07/2023] Open
Abstract
Hypoxia promotes an aggressive tumor phenotype with increased genomic instability, partially due to downregulation of DNA repair pathways. However, genome stability is also surveilled by cell cycle checkpoints. An important issue is therefore whether hypoxia also can influence the DNA damage-induced cell cycle checkpoints. Here, we show that hypoxia (24 h 0.2% O2) alters the expression of several G2 checkpoint regulators, as examined by microarray gene expression analysis and immunoblotting of U2OS cells. While some of the changes reflected hypoxia-induced inhibition of cell cycle progression, the levels of several G2 checkpoint regulators, in particular Cyclin B, were reduced in G2 phase cells after hypoxic exposure, as shown by flow cytometric barcoding analysis of individual cells. These effects were accompanied by decreased phosphorylation of a Cyclin dependent kinase (CDK) target in G2 phase cells after hypoxia, suggesting decreased CDK activity. Furthermore, cells pre-exposed to hypoxia showed increased G2 checkpoint arrest upon treatment with ionizing radiation. Similar results were found following other hypoxic conditions (∼0.03% O2 20 h and 0.2% O2 72 h). These results demonstrate that the DNA damage-induced G2 checkpoint can be altered as a consequence of hypoxia, and we propose that such alterations may influence the genome stability of hypoxic tumors.
Collapse
Affiliation(s)
- Grete Hasvold
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Christin Lund-Andersen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Malin Lando
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Sebastian Patzke
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Sissel Hauge
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - ZhenHe Suo
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Heidi Lyng
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Randi G Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway.
| |
Collapse
|
26
|
Lv P, Wang Y, Ma J, Wang Z, Li JL, Hong CS, Zhuang Z, Zeng YX. Inhibition of protein phosphatase 2A with a small molecule LB100 radiosensitizes nasopharyngeal carcinoma xenografts by inducing mitotic catastrophe and blocking DNA damage repair. Oncotarget 2015; 5:7512-24. [PMID: 25245035 PMCID: PMC4202140 DOI: 10.18632/oncotarget.2258] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC), while uncommon worldwide, is a major health problem in China. Although local radiation and surgery provide good control of NPC, better treatments that permit reductions in radiation dosing are needed. Inhibition of protein phosphatase 2A (PP2A), a ubiquitous multifunctional enzyme with critical roles in cell cycle regulation and DNA-damage response, reportedly sensitizes cancer cells to radiation and chemotherapy. We studied PP2A inhibition with LB100, a small molecule currently in a Phase I clinical trial, on radiosensitization of two human nasopharyngeal cell lines: CNE1, which is reportedly radioresistant, and CNE2. In both cell lines, LB100 exposure increased intracellular p-Plk1, TCTP, and Cdk1 and decreased p53, changes associated with cell cycle arrest, mitotic catastrophe and radio-inhibition of cell proliferation. Mice bearing subcutaneous xenografts of either cell line were administered 1.5 mg/kg LB100 daily for three days and a single dose of 20 Gy radiation (day 3), which produced marked and prolonged tumor mass regression (dose enhancement factors of 2.98 and 2.27 for CNE1 and CNE2 xenografts, respectively). Treatment with either LB100 or radiation alone only transiently inhibited xenograft growth. Our results support further exploration of PP2A inhibition as part of radiotherapy regimens for NPC and potentially other solid tumors.
Collapse
Affiliation(s)
- Peng Lv
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences (CAMS) ,Beijing , People's Republic of China. Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Yue Wang
- Institute for Medical Device Standardization Administration, National Institutes for Food and Drug Control, Beijing , People's Republic of China
| | - Jie Ma
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences (CAMS) ,Beijing , People's Republic of China
| | - Zheng Wang
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences (CAMS) ,Beijing , People's Republic of China
| | - Jing-Li Li
- Institute for Medical Device Standardization Administration, National Institutes for Food and Drug Control, Beijing , People's Republic of China
| | - Christopher S Hong
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD ,USA
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD ,USA
| | - Yi-Xin Zeng
- Cancer Institute and Hospital, Chinese Academy of Medical Sciences (CAMS) ,Beijing , People's Republic of China. Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong , People's Republic of China. State Key Laboratory of Oncology in Southern China, Guangzhou, Guangdong , People's Republic of China
| |
Collapse
|
27
|
Gordon IK, Lu J, Graves CA, Huntoon K, Frerich JM, Hanson RH, Wang X, Hong CS, Ho W, Feldman MJ, Ikejiri B, Bisht K, Chen XS, Tandle A, Yang C, Arscott WT, Ye D, Heiss JD, Lonser RR, Camphausen K, Zhuang Z. Protein Phosphatase 2A Inhibition with LB100 Enhances Radiation-Induced Mitotic Catastrophe and Tumor Growth Delay in Glioblastoma. Mol Cancer Ther 2015; 14:1540-1547. [PMID: 25939762 DOI: 10.1158/1535-7163.mct-14-0614] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 04/28/2015] [Indexed: 11/16/2022]
Abstract
Protein phosphatase 2A (PP2A) is a tumor suppressor whose function is lost in many cancers. An emerging, though counterintuitive, therapeutic approach is inhibition of PP2A to drive damaged cells through the cell cycle, sensitizing them to radiotherapy. We investigated the effects of PP2A inhibition on U251 glioblastoma cells following radiation treatment in vitro and in a xenograft mouse model in vivo. Radiotherapy alone augmented PP2A activity, though this was significantly attenuated with combination LB100 treatment. LB100 treatment yielded a radiation dose enhancement factor of 1.45 and increased the rate of postradiation mitotic catastrophe at 72 and 96 hours. Glioblastoma cells treated with combination LB100 and radiotherapy maintained increased γ-H2AX expression at 24 hours, diminishing cellular repair of radiation-induced DNA double-strand breaks. Combination therapy significantly enhanced tumor growth delay and mouse survival and decreased p53 expression 3.68-fold, compared with radiotherapy alone. LB100 treatment effectively inhibited PP2A activity and enhanced U251 glioblastoma radiosensitivity in vitro and in vivo. Combination treatment with LB100 and radiation significantly delayed tumor growth, prolonging survival. The mechanism of radiosensitization appears to be related to increased mitotic catastrophe, decreased capacity for repair of DNA double-strand breaks, and diminished p53 DNA-damage response pathway activity.
Collapse
Affiliation(s)
- Ira K Gordon
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jie Lu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - Christian A Graves
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kristin Huntoon
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Jason M Frerich
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Ryan H Hanson
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xiaoping Wang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Christopher S Hong
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Winson Ho
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Michael J Feldman
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Barbara Ikejiri
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Kheem Bisht
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xiaoyuan S Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - Anita Tandle
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Chunzhang Yang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - W Tristram Arscott
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Donald Ye
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Russell R Lonser
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
28
|
Wang H, Zhang X, Teng L, Legerski RJ. DNA damage checkpoint recovery and cancer development. Exp Cell Res 2015; 334:350-8. [PMID: 25842165 DOI: 10.1016/j.yexcr.2015.03.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/12/2015] [Accepted: 03/14/2015] [Indexed: 12/21/2022]
Abstract
Cell cycle checkpoints were initially presumed to function as a regulator of cell cycle machinery in response to different genotoxic stresses, and later found to play an important role in the process of tumorigenesis by acting as a guard against DNA over-replication. As a counterpart of checkpoint activation, the checkpoint recovery machinery is working in opposition, aiming to reverse the checkpoint activation and resume the normal cell cycle. The DNA damage response (DDR) and oncogene induced senescence (OIS) are frequently found in precancerous lesions, and believed to constitute a barrier to tumorigenesis, however, the DDR and OIS have been observed to be diminished in advanced cancers of most tissue origins. These findings suggest that when progressing from pre-neoplastic lesions to cancer, DNA damage checkpoint barriers are overridden. How the DDR checkpoint is bypassed in this process remains largely unknown. Activated cytokine and growth factor-signaling pathways were very recently shown to suppress the DDR and to promote uncontrolled cell proliferation in the context of oncovirus infection. In recent decades, data from cell line and tumor models showed that a group of checkpoint recovery proteins function in promoting tumor progression; data from patient samples also showed overexpression of checkpoint recovery proteins in human cancer tissues and a correlation with patients׳ poor prognosis. In this review, the known cell cycle checkpoint recovery proteins and their roles in DNA damage checkpoint recovery are reviewed, as well as their implications in cancer development. This review also provides insight into the mechanism by which the DDR suppresses oncogene-driven tumorigenesis and tumor progression.
Collapse
Affiliation(s)
- Haiyong Wang
- First affiliated hospital, Zhejiang University, School of medicine, Cancer Center, 79 Qingchun Road, Hangzhou 310003, China
| | - Xiaoshan Zhang
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Department of Genetics Unit 1010, 1515 Holcombe Blvd. Houston, TX 77030 USA
| | - Lisong Teng
- First affiliated hospital, Zhejiang University, School of medicine, Cancer Center, 79 Qingchun Road, Hangzhou 310003, China.
| | - Randy J Legerski
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Department of Genetics Unit 1010, 1515 Holcombe Blvd. Houston, TX 77030 USA.
| |
Collapse
|
29
|
Zhu DW, Yuan YX, Qiao JK, Yu C, Yang X, Wang LZ, Zhang ZY, Zhong LP. Enhanced anticancer activity of a protein phosphatase 2A inhibitor on chemotherapy and radiation in head and neck squamous cell carcinoma. Cancer Lett 2014; 356:773-80. [PMID: 25449438 DOI: 10.1016/j.canlet.2014.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 11/18/2022]
Abstract
The aim of this study is to eliminate more cancer cells by promoting them from quiescence into cell cycle or by changing their molecular events, leading them to be sensitive to radiation or chemotherapy. Protein phosphatase 2A plays an important role in many cellular functions and regulates various biological processes. It is unclear that LB1, which is an inhibitor of protein phosphatase 2A, has enhanced anticancer activity on chemotherapy (cisplatin and 5-fluorourcil) and radiation in head and neck squamous cell carcinoma (HNSCC). Herein, we performed both in vitro and in vivo studies to determine the anticancer activity of LB1 on chemotherapy and radiation in HNSCC, with detection of p53 expression, AKT and MDM2 phosphorylation. In vitro studies indicated that, LB1 could significantly enhance the cytotoxicity of cisplatin, 5-fluorourcil, and radiation; LB1 could also significantly enhance the treatment effect of cisplatin in nude mice. The anticancer activity of LB1 was mediated by increased AKT phosphorylation and decreased p53 expression with increased MDM2 phosphorylation, especially when combined with cisplatin. Our data suggest a strategy of improving treatment effect through the enhanced anticancer activity of LB1 on cisplatin-based chemotherapy and radiation in HNSCC.
Collapse
Affiliation(s)
- Dong-wang Zhu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-xiang Yuan
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-ke Qiao
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Yu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Yang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-zhen Wang
- Department of Oral Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-yuan Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lai-ping Zhong
- Department of Oral & Maxillofacial-Head & Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
30
|
Ling H, Lu LF, He J, Xiao GH, Jiang H, Su Q. Diallyl disulfide selectively causes checkpoint kinase-1 mediated G2/M arrest in human MGC803 gastric cancer cell line. Oncol Rep 2014; 32:2274-82. [PMID: 25176258 DOI: 10.3892/or.2014.3417] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/04/2014] [Indexed: 11/06/2022] Open
Abstract
Previous studies have shown that diallyl disulfide (DADS), a naturally occurring anticancer agent in garlic, arrested human gastric cancer cells (MGC803) in the G2/M phase of the cell cycle. Due to the importance of cell cycle redistribution in DADS-mediated anticarcinogenic effects, we investigated the role of checkpoint kinases (Chk1 and Chk2) during DADS-induced cell cycle arrest. In the present study, the northern blot analysis showed that mRNA expression of for Chkl and Chk2 was unchanged. Notably, DADS induced the accumulation of phosphorylated Chk1, but not of Chk2, activated phospho-ATR (ATM-RAD3-related gene), and dowregulated CDC25C and cyclin B1 expression. Furthermore, CDC25C was immunoprecipitated by anti-Chk1 but not anti-Chk2. Results of the overexpression and knockdown studies, showed that Chk1 but not Chk2 regulated the DADS-induced G2/M arrest of MGC803 cells. The overexpression of Chk1 resulted in significantly increased DADS-induced G2/M arrest, increased DADS-induced Chk1 phosphorylation and inhibited CDC25C expression. Knockdown of Chk1 reduced DADS‑induced G2/M arrest and blocked the DADS-induced inhibition of CDC25C and cyclin B1 expression. These results suggested that Chk1 is important in DADS‑induced cell cycle G2/M arrest in the human MGC803 gastric cancer cell line. Furthermore, the DADS-induced G2/M checkpoint response is mediated by Chk1 signaling through ATR/Chk1/CDC25C/cyclin B1.
Collapse
Affiliation(s)
- Hui Ling
- Key Laboratory of Tumor Cellular and Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, P.R. China
| | - Li-Feng Lu
- Key Laboratory of Tumor Cellular and Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, P.R. China
| | - Jie He
- Key Laboratory of Tumor Cellular and Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, P.R. China
| | - Guo-Hua Xiao
- Key Laboratory of Tumor Cellular and Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, P.R. China
| | - Hao Jiang
- Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qi Su
- Key Laboratory of Tumor Cellular and Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
31
|
A novel function of HER2/Neu in the activation of G2/M checkpoint in response to γ-irradiation. Oncogene 2014; 34:2215-26. [PMID: 24909175 DOI: 10.1038/onc.2014.167] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 04/21/2014] [Accepted: 05/06/2014] [Indexed: 12/13/2022]
Abstract
In response to γ-irradiation (IR)-induced DNA damage, activation of cell cycle checkpoints results in cell cycle arrest, allowing time for DNA repair before cell cycle re-entry. Human cells contain G1 and G2 cell cycle checkpoints. While G1 checkpoint is defective in most cancer cells, commonly due to mutations and/or alterations in the key regulators of G1 checkpoint (for example, p53, cyclin D), G2 checkpoint is rarely impaired in cancer cells, which is important for cancer cell survival. G2 checkpoint activation involves activation of ataxia telangiectasia-mutated (ATM)/ATM- and rad3-related (ATR) signalings, which leads to the inhibition of Cdc2 kinase and subsequent G2/M cell cycle arrest. Previous studies from our laboratory show that G2 checkpoint activation following IR exposure of MCF-7 breast cancer cells is dependent on the activation of extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) signaling. As HER receptor tyrosine kinases (RTKs), which have important roles in cell proliferation and survival, have been shown to activate ERK1/2 signaling in response to various stimuli, we investigated the role of HER RTKs in IR-induced G2/M checkpoint response in breast cancer cells. Results of the present studies indicate that IR exposure resulted in a striking increase in the phosphorylation of HER1, HER2, HER3 and HER4 in MCF-7 cells, indicative of activation of these proteins. Furthermore, specific inhibition of HER2 using an inhibitor, short hairpin RNA and dominant-negative mutant HER2 abolished IR-induced activation of ATM/ATR signaling, phosphorylation of Cdc2-Y15 and subsequent induction of G2/M arrest. Moreover, the inhibition of HER2 also abrogated IR-induced ERK1/2 phosphorylation. In contrast, inhibition of HER1 using specific inhibitors or decreasing expression of HER3 or HER4 using short hairpin RNAs did not block the induction of G2/M arrest following IR. These results suggest an important role of HER2 in the activation of G2/M checkpoint response following IR.
Collapse
|
32
|
Telomerase activation as a repair response to radiation-induced DNA damage in Y79 retinoblastoma cells. Cancer Lett 2013; 340:82-7. [DOI: 10.1016/j.canlet.2013.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 06/11/2013] [Accepted: 07/05/2013] [Indexed: 01/09/2023]
|
33
|
Hu Z, Kong F, Si M, Tian K, Yu LX, Young CYF, Yuan H, Lou H. Riccardin D Exerts Its Antitumor Activity by Inducing DNA Damage in PC-3 Prostate Cancer Cells In Vitro and In Vivo. PLoS One 2013; 8:e74387. [PMID: 24069304 PMCID: PMC3775815 DOI: 10.1371/journal.pone.0074387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 07/31/2013] [Indexed: 11/19/2022] Open
Abstract
We recently reported that Riccardin D (RD) was able to induce apoptosis by targeting Topo II. Here, we found that RD induced cell cycle arrest in G2/M phase in PC-3 cells, and caused remarkable DNA damage as evidenced by induction of γH2AX foci, micronuclei, and DNA fragmentation in Comet assay. Time kinetic and dose-dependent studies showed that ATM/Chk2 and ATR/Chk1 signaling pathways were sequentially activated in response to RD. Blockage of ATM/ATR signaling led to the attenuation of RD-induced γH2AX, and to the partial recovery of cell proliferation. Furthermore, RD exposure resulted in the inactivation of BRCA1, suppression of HR and NHEJ repair activity, and downregulation of the expressions and DNA-end binding activities of Ku70/86. Consistent with the observations, microarray data displayed that RD triggered the changes in genes responsible for cell proliferation, cell cycle, DNA damage and repair, and apoptosis. Administration of RD to xenograft mice reduced tumor growth, and coordinately caused alterations in the expression of genes involved in DNA damage and repair, along with cell apoptosis. Thus, this finding identified a novel mechanism by which RD affects DNA repair and acts as a DNA damage agent in prostate cancer.
Collapse
Affiliation(s)
- Zhongyi Hu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
- Department of Natural Product Chemistry, Shandong University School of Pharmaceutical Sciences, Jinan, China
| | - Feng Kong
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Manfei Si
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Keli Tian
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Lin Xi Yu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Charles Y. F. Young
- Department of Urology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Huiqing Yuan
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
- * E-mail: (HY); (HL)
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Shandong University School of Pharmaceutical Sciences, Jinan, China
- * E-mail: (HY); (HL)
| |
Collapse
|
34
|
Tian Y, Xie Q, Tian Y, Liu Y, Huang Z, Fan C, Hou B, Sun D, Yao K, Chen T. Radioactive ¹²⁵I seed inhibits the cell growth, migration, and invasion of nasopharyngeal carcinoma by triggering DNA damage and inactivating VEGF-A/ERK signaling. PLoS One 2013; 8:e74038. [PMID: 24040157 PMCID: PMC3769370 DOI: 10.1371/journal.pone.0074038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/25/2013] [Indexed: 11/19/2022] Open
Abstract
Although radiotherapy technology has progressed rapidly in the past decade, the inefficiency of radiation and cancer cell resistance mean that the 5-year survival rate of patients with nasopharyngeal carcinoma (NPC) is low. Radioactive 125I seed implantation has received increasing attention as a clinical treatment for cancers. Vascular endothelial growth factor-A (VEGF-A) is one of the most important members of the VEGF family and plays an important role in cell migration through the extracellular-signal-regulated kinase (ERK) pathway. Here we show that radioactive 125I seeds more effectively inhibit NPC cell growth through DNA damage and subsequent induction of apoptosis, compared with X-ray irradiation. Moreover, cell migration was effectively inhibited by 125I seed irradiation through VEGF-A/ERK inactivation. VEGF-A pretreatment significantly blocked 125I seed irradiation-induced inhibition of cell migration by recovering the levels of phosphorylated ERK (p-ERK) protein. Interestingly, in vivo study results confirmed that 125I seed irradiation was more effective in inhibiting tumor growth than X-ray irradiation. Taken together, these results suggest that radioactive 125I seeds exert novel anticancer activity by triggering DNA damage and inactivating VEGF-A/ERK signaling. Our finding provides evidence for the efficacy of 125I seeds for treating NPC patients, especially those with local recurrence.
Collapse
Affiliation(s)
- Yunhong Tian
- Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Oncology, Armed Police Hospital of Guangdong Province, Guangzhou, Guangdong Province, People's Republic of China
| | - Qiang Xie
- Department of Oncology, Armed Police Hospital of Guangdong Province, Guangzhou, Guangdong Province, People's Republic of China
- Department of Pathology, Medical College of Jinan University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yunming Tian
- State Key Laboratory Oncology in Southern China, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Radiation Oncology, Cancer Center of Sun Yat-Sen University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Ying Liu
- Department of Oncology, Armed Police Hospital of Guangdong Province, Guangzhou, Guangdong Province, People's Republic of China
| | - Zuoping Huang
- Department of Oncology, Armed Police Hospital of Guangdong Province, Guangzhou, Guangdong Province, People's Republic of China
| | - Cundong Fan
- Department of Chemistry, Jinan University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Bing Hou
- Department of Oncology, Armed Police Hospital of Guangdong Province, Guangzhou, Guangdong Province, People's Republic of China
| | - Dan Sun
- Department of Oncology, Armed Police Hospital of Guangdong Province, Guangzhou, Guangdong Province, People's Republic of China
| | - Kaitai Yao
- Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- * E-mail: (KY), (TC)
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, Guangdong Province, People’s Republic of China
- * E-mail: (KY), (TC)
| |
Collapse
|
35
|
Wei D, Parsels LA, Karnak D, Davis MA, Parsels JD, Marsh AC, Zhao L, Maybaum J, Lawrence TS, Sun Y, Morgan MA. Inhibition of protein phosphatase 2A radiosensitizes pancreatic cancers by modulating CDC25C/CDK1 and homologous recombination repair. Clin Cancer Res 2013; 19:4422-32. [PMID: 23780887 DOI: 10.1158/1078-0432.ccr-13-0788] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To identify targets whose inhibition may enhance the efficacy of chemoradiation in pancreatic cancer and thus improve survival, we conducted an siRNA library screen in pancreatic cancer cells. We investigated PPP2R1A, a scaffolding subunit of protein phosphatase 2A (PP2A) as a lead radiosensitizing target. EXPERIMENTAL DESIGN We determined the effect of PP2A inhibition by genetic (PPP2R1A siRNA) and pharmacologic (LB100, a small molecule entering phase I clinical trials) approaches on radiosensitization of Panc-1 and MiaPaCa-2 pancreatic cancer cells both in vitro and in vivo. RESULTS PPP2R1A depletion by siRNA radiosensitized Panc-1 and MiaPaCa-2 cells, with radiation enhancement ratios of 1.4 (P < 0.05). Likewise, LB100 produced similar radiosensitization in pancreatic cancer cells, but minimal radiosensitization in normal small intestinal cells. Mechanistically, PPP2R1A siRNA or LB100 caused aberrant CDK1 activation, likely resulting from accumulation of the active forms of PLK1 (pPLK1 T210) and CDC25C (pCDC25C T130). Furthermore, LB100 inhibited radiation-induced Rad51 focus formation and homologous recombination repair (HRR), ultimately leading to persistent radiation-induced DNA damage, as reflected by γ-H2AX expression. Finally, we identified CDC25C as a key PP2A substrate involved in LB100-mediated radiosensitization as depletion of CDC25C partially reversed LB100-mediated radiosensitization. In a mouse xenograft model of human pancreatic cancer, LB100 produced significant radiosensitization with minimal weight loss. CONCLUSIONS Collectively, our data show that PP2A inhibition radiosensitizes pancreatic cancer both in vitro and in vivo via activation of CDC25C/CDK1 and inhibition of HRR, and provide proof-of-concept evidence that PP2A is a promising target for the improvement of local therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Dongping Wei
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan 48109-5637, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kaufmann WK, Anderson CW. Compensation, crosstalk and sequestering: the currency of checkpoints in cancer. Cell Cycle 2013; 12:1163-4. [PMID: 23549170 PMCID: PMC3674080 DOI: 10.4161/cc.24486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- William K Kaufmann
- Department of Pathology and Laboratory of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
37
|
Palii SS, Cui Y, Innes CL, Paules RS. Dissecting cellular responses to irradiation via targeted disruptions of the ATM-CHK1-PP2A circuit. Cell Cycle 2013; 12:1105-18. [PMID: 23462183 DOI: 10.4161/cc.24127] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Exposure of proliferating cells to genotoxic stresses activates a cascade of signaling events termed the DNA damage response (DDR). The DDR preserves genetic stability by detecting DNA lesions, activating cell cycle checkpoints and promoting DNA damage repair. The phosphoinositide 3-kinase-related kinases (PIKKs) ataxia telangiectasia-mutated (ATM), ATM and Rad 3-related kinase (ATR) and DNA-dependent protein kinase (DNA-PK) are crucial for sensing lesions and signal transduction. The checkpoint kinase 1 (CHK1) is a traditional ATR target involved in DDR and normal cell cycle progression and represents a pharmacological target for anticancer regimens. This study employed cell lines stably depleted for CHK1, ATM or both for dissecting cross-talk and compensatory effects on G(2)/M checkpoint in response to ionizing radiation (IR). We show that a 90% depletion of CHK1 renders cells radiosensitive without abrogating their IR-mediated G(2)/M checkpoint arrest. ATM phosphorylation is enhanced in CHK1-deficient cells compared with their wild-type counterparts. This correlates with lower nuclear abundance of the PP2A catalytic subunit in CHK1-depleted cells. Stable depletion of CHK1 in an ATM-deficient background showed only a 50% reduction from wild-type CHK1 protein expression levels and resulted in an additive attenuation of the G(2)/M checkpoint response compared with the individual knockdowns. ATM inhibition and 90% CHK1 depletion abrogated the early G(2)/M checkpoint and precluded the cells from mounting an efficient compensatory response to IR at later time points. Our data indicates that dual targeting of ATM and CHK1 functionalities disrupts the compensatory response to DNA damage and could be exploited for developing efficient anti-neoplastic treatments.
Collapse
Affiliation(s)
- Stela S Palii
- Environmental Stress and Cancer Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | | | | | | |
Collapse
|
38
|
Shimada M, Nakanishi M. Response to DNA damage: why do we need to focus on protein phosphatases? Front Oncol 2013; 3:8. [PMID: 23386996 PMCID: PMC3560363 DOI: 10.3389/fonc.2013.00008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 01/09/2013] [Indexed: 01/07/2023] Open
Abstract
Eukaryotic cells are continuously threatened by unavoidable errors during normal DNA replication or various sources of genotoxic stresses that cause DNA damage or stalled replication. To maintain genomic integrity, cells have developed a coordinated signaling network, known as the DNA damage response (DDR). Following DNA damage, sensor molecules detect the presence of DNA damage and transmit signals to downstream transducer molecules. This in turn conveys the signals to numerous effectors, which initiate a large number of specific biological responses, including transient cell cycle arrest mediated by checkpoints, DNA repair, and apoptosis. It is recently becoming clear that dephosphorylation events are involved in keeping DDR factors inactive during normal cell growth. Moreover, dephosphorylation is required to shut off checkpoint arrest following DNA damage and has been implicated in the activation of the DDR. Spatial and temporal regulation of phosphorylation events is essential for the DDR, and fine-tuning of phosphorylation is partly mediated by protein phosphatases. While the role of kinases in the DDR has been well documented, the complex roles of protein dephosphorylation have only recently begun to be investigated. Therefore, it is important to focus on the role of phosphatases and to determine how their activity is regulated upon DNA damage. In this work, we summarize current knowledge on the involvement of serine/threonine phosphatases, especially the protein phosphatase 1, protein phosphatase 2A, and protein phosphatase Mg2+/Mn2+-dependent families, in the DDR.
Collapse
Affiliation(s)
- Midori Shimada
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University Nagoya, Japan
| | | |
Collapse
|
39
|
Kalev P, Simicek M, Vazquez I, Munck S, Chen L, Soin T, Danda N, Chen W, Sablina A. Loss of PPP2R2A inhibits homologous recombination DNA repair and predicts tumor sensitivity to PARP inhibition. Cancer Res 2012; 72:6414-24. [PMID: 23087057 DOI: 10.1158/0008-5472.can-12-1667] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reversible phosphorylation plays a critical role in DNA repair. Here, we report the results of a loss-of-function screen that identifies the PP2A heterotrimeric serine/threonine phosphatases PPP2R2A, PPP2R2D, PPP2R5A, and PPP2R3C in double-strand break (DSB) repair. In particular, we found that PPP2R2A-containing complexes directly dephosphorylated ATM at S367, S1893, and S1981 to regulate its retention at DSB sites. Increased ATM phosphorylation triggered by PPP2R2A attenuation dramatically upregulated the activity of the downstream effector kinase CHK2, resulting in G(1) to S-phase cell-cycle arrest and downregulation of BRCA1 and RAD51. In tumor cells, blocking PPP2R2A thereby impaired the high-fidelity homologous recombination repair pathway and sensitized cells to small-molecule inhibitors of PARP. We found that PPP2R2A was commonly downregulated in non-small cell lung carcinomas, suggesting that PPP2R2A status may serve as a marker to predict therapeutic efficacy to PARP inhibition. In summary, our results deepen understanding of the role of PP2A family phosphatases in DNA repair and suggest PPP2R2A as a marker for PARP inhibitor responses in clinic.
Collapse
Affiliation(s)
- Peter Kalev
- VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schnerch D, Yalcintepe J, Schmidts A, Becker H, Follo M, Engelhardt M, Wäsch R. Cell cycle control in acute myeloid leukemia. Am J Cancer Res 2012; 2:508-528. [PMID: 22957304 PMCID: PMC3433102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/27/2012] [Indexed: 06/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is the result of a multistep transforming process of hematopoietic precursor cells (HPCs) which enables them to proceed through limitless numbers of cell cycles and to become resistant to cell death. Increased proliferation renders these cells vulnerable to acquiring mutations and may favor leukemic transformation. Here, we review how deregulated cell cycle control contributes to increased proliferation in AML and favors genomic instability, a prerequisite to confer selective advantages to particular clones in order to adapt and independently proliferate in the presence of a changing microenvironment. We discuss the connection between differentiation and proliferation with regard to leukemogenesis and outline the impact of specific alterations on response to therapy. Finally, we present examples, how a better understanding of cell cycle regulation and deregulation has already led to new promising therapeutic strategies.
Collapse
Affiliation(s)
- Dominik Schnerch
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Thompson LH. Recognition, signaling, and repair of DNA double-strand breaks produced by ionizing radiation in mammalian cells: the molecular choreography. Mutat Res 2012; 751:158-246. [PMID: 22743550 DOI: 10.1016/j.mrrev.2012.06.002] [Citation(s) in RCA: 261] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 06/09/2012] [Accepted: 06/16/2012] [Indexed: 12/15/2022]
Abstract
The faithful maintenance of chromosome continuity in human cells during DNA replication and repair is critical for preventing the conversion of normal diploid cells to an oncogenic state. The evolution of higher eukaryotic cells endowed them with a large genetic investment in the molecular machinery that ensures chromosome stability. In mammalian and other vertebrate cells, the elimination of double-strand breaks with minimal nucleotide sequence change involves the spatiotemporal orchestration of a seemingly endless number of proteins ranging in their action from the nucleotide level to nucleosome organization and chromosome architecture. DNA DSBs trigger a myriad of post-translational modifications that alter catalytic activities and the specificity of protein interactions: phosphorylation, acetylation, methylation, ubiquitylation, and SUMOylation, followed by the reversal of these changes as repair is completed. "Superfluous" protein recruitment to damage sites, functional redundancy, and alternative pathways ensure that DSB repair is extremely efficient, both quantitatively and qualitatively. This review strives to integrate the information about the molecular mechanisms of DSB repair that has emerged over the last two decades with a focus on DSBs produced by the prototype agent ionizing radiation (IR). The exponential growth of molecular studies, heavily driven by RNA knockdown technology, now reveals an outline of how many key protein players in genome stability and cancer biology perform their interwoven tasks, e.g. ATM, ATR, DNA-PK, Chk1, Chk2, PARP1/2/3, 53BP1, BRCA1, BRCA2, BLM, RAD51, and the MRE11-RAD50-NBS1 complex. Thus, the nature of the intricate coordination of repair processes with cell cycle progression is becoming apparent. This review also links molecular abnormalities to cellular pathology as much a possible and provides a framework of temporal relationships.
Collapse
Affiliation(s)
- Larry H Thompson
- Biology & Biotechnology Division, L452, Lawrence Livermore National Laboratory, P.O. Box 808, Livermore, CA 94551-0808, United States.
| |
Collapse
|
42
|
Bhoopathi P, Gorantla B, Sailaja GS, Gondi CS, Gujrati M, Klopfenstein JD, Rao JS. SPARC overexpression inhibits cell proliferation in neuroblastoma and is partly mediated by tumor suppressor protein PTEN and AKT. PLoS One 2012; 7:e36093. [PMID: 22567126 PMCID: PMC3342296 DOI: 10.1371/journal.pone.0036093] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/29/2012] [Indexed: 01/01/2023] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is also known as BM-40 or Osteonectin, a multi-functional protein modulating cell–cell and cell–matrix interactions. In cancer, SPARC is not only linked with a highly aggressive phenotype, but it also acts as a tumor suppressor. In the present study, we sought to characterize the function of SPARC and its role in sensitizing neuroblastoma cells to radio-therapy. SPARC overexpression in neuroblastoma cells inhibited cell proliferation in vitro. Additionally, SPARC overexpression significantly suppressed the activity of AKT and this suppression was accompanied by an increase in the tumor suppressor protein PTEN both in vitro and in vivo. Restoration of neuroblastoma cell radio-sensitivity was achieved by overexpression of SPARC in neuroblastoma cells in vitro and in vivo. To confirm the role of the AKT in proliferation inhibited by SPARC overexpression, we transfected neuroblastoma cells with a plasmid vector carrying myr-AKT. Myr-AKT overexpression reversed SPARC-mediated PTEN and increased proliferation of neuroblastoma cells in vitro. PTEN overexpression in parallel with SPARC siRNA resulted in decreased AKT phosphorylation and proliferation in vitro. Taken together, these results establish SPARC as an effector of AKT-PTEN-mediated inhibition of proliferation in neuroblastoma in vitro and in vivo.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Bharathi Gorantla
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - G. S. Sailaja
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Christopher S. Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Meena Gujrati
- Department of Pathology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Jeffrey D. Klopfenstein
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
- * E-mail:
| |
Collapse
|
43
|
del Carmen MG, Birrer M, Schorge JO. Clear cell carcinoma of the ovary: a review of the literature. Gynecol Oncol 2012; 126:481-90. [PMID: 22525820 DOI: 10.1016/j.ygyno.2012.04.021] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/12/2012] [Accepted: 04/16/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Different histologic types of epithelial ovarian cancer may represent different diseases with unique clinical and molecular characteristics. Clear cell carcinoma (CCC) of the ovary has been reported as having a worse prognosis than high grade serous epithelial ovarian cancer (EOC). This article critically reviews the literature pertinent to the pathology, pathogenesis, diagnosis, management, and outcome of patients with ovarian CCC. METHODS MEDLINE was searched for all research articles published in English between January 01, 1977 and January 30, 2012 which reported on patients diagnosed with ovarian CCC. Given the rarity of this tumor, studies were not limited by design or number of reported patients. RESULTS Ovarian CCC tumors represent 5-25% of ovarian cancers. Its histologic diagnosis can be challenging, resulting often times in misclassification of these tumors. Ovarian CCC tends to present at earlier stages and has been associated with endometriosis, ARID1A and PIK3CA mutations. When compared to stage-matched controls, patients with early-stage ovarian CCCs may have a better prognosis than patients with high-grade serous tumors. For those with advanced stage disease, high-grade serous histology confers a better prognosis than ovarian CCC. Patients with Stage IC-IV have a relatively poor prognosis and efforts should center in discovery of more effective treatment strategies. CONCLUSIONS Ovarian CCC is a biologically distinct entity, different from high-grade serous EOC. Future studies should explore the role of targeted therapies in the management of ovarian CCC.
Collapse
Affiliation(s)
- Marcela G del Carmen
- Division of Gynecologic Oncology, Vincent Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | |
Collapse
|
44
|
Su YQ, Sugiura K, Sun F, Pendola JK, Cox GA, Handel MA, Schimenti JC, Eppig JJ. MARF1 regulates essential oogenic processes in mice. Science 2012; 335:1496-9. [PMID: 22442484 DOI: 10.1126/science.1214680] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Development of fertilization-competent oocytes depends on integrated processes controlling meiosis, cytoplasmic development, and maintenance of genomic integrity. We show that meiosis arrest female 1 (MARF1) is required for these processes in mammalian oocytes. Mutations of Marf1 cause female infertility characterized by up-regulation of a cohort of transcripts, increased retrotransposon expression, defective cytoplasmic maturation, and meiotic arrest. Up-regulation of protein phosphatase 2 catalytic subunit (PPP2CB) is key to the meiotic arrest phenotype. Moreover, Iap and Line1 retrotransposon messenger RNAs are also up-regulated, and, concomitantly, DNA double-strand breaks are elevated in mutant oocytes. Therefore MARF1, by suppressing levels of specific transcripts, is an essential regulator of important oogenic processes leading to female fertility and the development of healthy offspring.
Collapse
Affiliation(s)
- You-Qiang Su
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hofstetter CP, Burkhardt JK, Shin BJ, Gürsel DB, Mubita L, Gorrepati R, Brennan C, Holland EC, Boockvar JA. Protein phosphatase 2A mediates dormancy of glioblastoma multiforme-derived tumor stem-like cells during hypoxia. PLoS One 2012; 7:e30059. [PMID: 22253878 PMCID: PMC3256196 DOI: 10.1371/journal.pone.0030059] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 12/13/2011] [Indexed: 11/19/2022] Open
Abstract
PURPOSE The hypoxic microenvironment of glioblastoma multiforme (GBM) is thought to increase resistance to cancer therapies. Recent evidence suggests that hypoxia induces protein phosphatase 2A (PP2A), a regulator of cell cycle and cell death. The effects of PP2A on GBM tumor cell proliferation and survival during hypoxic conditions have not been studied. EXPERIMENTAL DESIGN Expression of PP2A subunits and HIF-α proteins was measured in 65 high-grade astrocytoma and 18 non-neoplastic surgical brain specimens by western blotting. PP2A activity was measured by an immunoprecipitation assay. For in vitro experiments, GBM-derived tumor stem cell-like cells (TSCs) were exposed to severe hypoxia produced by either CoCl₂ or 1% O₂. PP2A activity was inhibited either by okadaic acid or by shRNA depletion of the PP2A C subunit. Effects of PP2A activity on cell cycle progression and cell survival during hypoxic conditions were assessed using flow cytometry. RESULTS In our patient cohort, PP2A activity was positively correlated with HIF-1∝ protein expression (P = 0.002). Patients with PP2A activity levels above 160 pMP had significantly worse survival compared to patients with levels below this threshold (P = 0.002). PP2A activity was an independent predictor of survival on multivariable analysis (P = 0.009). In our in vitro experiments, we confirmed that severe hypoxia induces PP2A activity in TSCs 6 hours after onset of exposure. PP2A activity mediated G1/S phase growth inhibition and reduced cellular ATP consumption in hypoxic TSCs. Conversely, inhibition of PP2A activity led to increased cell proliferation, exhaustion of intracellular ATP, and accelerated P53-independent cell death of hypoxic TSCs. CONCLUSIONS Our results suggest that PP2A activity predicts poor survival in GBM. PP2A appears to reduce the metabolic demand of hypoxic TSCs and enhances tumor cell survival. Modulation of PP2A may be a potential target for cancer therapy.
Collapse
Affiliation(s)
- Christoph P Hofstetter
- Department of Neurological Surgery, Weill Cornell Brain Tumor Center, Weill Cornell Medical College, New York Presbyterian Hospital, New York, New York, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Lee DH, Chowdhury D. What goes on must come off: phosphatases gate-crash the DNA damage response. Trends Biochem Sci 2011; 36:569-77. [PMID: 21930385 DOI: 10.1016/j.tibs.2011.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/24/2022]
Abstract
DNA-damage-induced phospho-signaling has been studied for decades, with a focus mainly on initiation of the signaling cascade, and the kinases activated by DNA lesions. It is widely accepted that the balance of phosphorylation needs to be restored and/or maintained by phosphatases, yet there have only been sporadic efforts to investigate the impact of phosphatases on DNA repair. Recent advances in phosphoproteomic strategies and implementation of large genetic screens indicate that these enzymes play pivotal roles in these signaling networks. Dephosphorylation of repair proteins is crucial for efficient DNA repair, and the recommencement of cell division post-repair. Here, we focus on serine/threonine phosphatases implicated in dephosphorylation of DNA repair factors, summarizing recent findings and speculating on untested roles of phosphatases in the DNA damage response.
Collapse
Affiliation(s)
- Dong-Hyun Lee
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
48
|
Clear cell carcinoma of the ovary: a report from the first Ovarian Clear Cell Symposium, June 24th, 2010. Gynecol Oncol 2011; 121:407-15. [PMID: 21276610 DOI: 10.1016/j.ygyno.2011.01.005] [Citation(s) in RCA: 186] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/01/2011] [Accepted: 01/03/2011] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Recent literature has highlighted histological types of ovarian carcinoma as distinct diseases, each with unique clinical and molecular features. Historically, the diagnosis of ovarian clear cell carcinoma (CCC) has been of concern to both patients and physicians due to reports that CCC is associated with a worse prognosis than the more common serous type of ovarian carcinoma (HGSC). This review discusses the unique features of ovarian CCC. METHODS In June of 2010, a group of researchers and clinicians convened in Vancouver to review and discuss the clinical, pathological, molecular, and treatment-related features of CCC. RESULTS CCC is the second most common type of ovarian epithelial cancer, representing 5-25% of ovarian carcinomas. It is characterised by its association with endometriosis, and frequent mutations of ARID1A and PIK3CA. Low-stage CCC appears to have a better outcome than stage matched HGSC, while the opposite is true for high-stage disease, suggesting that the current standard treatments applied to HGSC are ineffective for CCC. CONCLUSIONS Ovarian CCC is highly distinct from HGSC, and a clearer understanding of the basic biology of this disease is needed. Alternative therapies should be explored: irradiation and targeting disease-specific molecular markers should be examined in greater detail. Finally, novel approaches to clinical trial design are needed due to the smaller numbers of patients affected.
Collapse
|
49
|
Abstract
The cellular response to DNA damage is a crucial surveillance mechanism that maintains genomic integrity and prevents cancer progression. Previous studies identified multiple Ser/Thr protein kinases that have pivotal roles in the activation of this response. It is interesting that a growing body of evidence suggests that these kinases and their substrates are under tight modulation by numerous Ser/Thr phosphatases. In this study, we review recent reports that reveal new functions and regulation of these phosphatases. Similar to the kinases in this pathway, phosphatases may also be intimately involved in cancer progression and present valuable targets for cancer therapy.
Collapse
|