1
|
Fan H, Xu Z, Yao K, Zheng B, Zhang Y, Wang X, Zhang T, Li X, Hu H, Yue B, Hu Z, Zheng H. Osteoclast Cancer Cell Metabolic Cross-talk Confers PARP Inhibitor Resistance in Bone Metastatic Breast Cancer. Cancer Res 2024; 84:449-467. [PMID: 38038966 DOI: 10.1158/0008-5472.can-23-1443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023]
Abstract
The majority of patients with late-stage breast cancer develop distal bone metastases. The bone microenvironment can affect response to therapy, and uncovering the underlying mechanisms could help identify improved strategies for treating bone metastatic breast cancer. Here, we observed that osteoclasts reduced the sensitivity of breast cancer cells to DNA damaging agents, including cisplatin and the PARP inhibitor (PARPi) olaparib. Metabolic profiling identified elevated glutamine production by osteoclasts. Glutamine supplementation enhanced the survival of breast cancer cells treated with DNA damaging agents, while blocking glutamine uptake increased sensitivity and suppressed bone metastasis. GPX4, the critical enzyme responsible for glutathione oxidation, was upregulated in cancer cells following PARPi treatment through stress-induced ATF4-dependent transcriptional programming. Increased glutamine uptake and GPX4 upregulation concertedly enhanced glutathione metabolism in cancer cells to help neutralize oxidative stress and generate PARPi resistance. Analysis of paired patient samples of primary breast tumors and bone metastases revealed significant induction of GPX4 in bone metastases. Combination therapy utilizing PARPi and zoledronate, which blocks osteoclast activity and thereby reduces the microenvironmental glutamine supply, generated a synergistic effect in reducing bone metastasis. These results identify a role for glutamine production by bone-resident cells in supporting metastatic cancer cells to overcome oxidative stress and develop resistance to DNA-damaging therapies. SIGNIFICANCE Metabolic interaction between osteoclasts and tumor cells contributes to resistance to DNA-damaging agents, which can be blocked by combination treatment with PARP and osteoclast inhibitors to reduce bone metastatic burden.
Collapse
Affiliation(s)
- Huijuan Fan
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Zhanao Xu
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Ke Yao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Bingxin Zheng
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yuan Zhang
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xuxiang Wang
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Tengjiang Zhang
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xuan Li
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Haitian Hu
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Bin Yue
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
2
|
Fu Y, Lv Z, Kong D, Fan Y, Dong B. High abundance of CDC45 inhibits cell proliferation through elevation of HSPA6. Cell Prolif 2022; 55:e13257. [PMID: 35642733 PMCID: PMC9251052 DOI: 10.1111/cpr.13257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES CDC45 is the core component of CMG (CDC45-MCMs-GINS) complex that plays important role in the initial step of DNA replication in eukaryotic cells. The expression level of cdc45 is under the critical control for the accurate cell cycle progression. Loss-of-function of cdc45 has been demonstrated to inhibit cell proliferation and leads to cell death due to the inhibition of DNA replication and G1-phase arrest. An increasing of CDC45 inhibits cell proliferation as well. Nevertheless, a systematic analysis of the effect of high dose of CDC45 on cell physiology and behaviors is unclear. In the present study, we aimed to investigate the effects and mechanisms of high dose of CDC45 on cell behaviors. MATERIALS AND METHODS We overexpressed cdc45 in cultured cell lines, Ciona and Drosophila embryos, respectively. The cell cycle progression was examined by the BrdU incorporation experiment, flow cytometry and PH3 (phospho-Histone 3) staining. RNA-sequencing analysis and qRT-PCR were carried out to screen the affected genes in HeLa cells overexpressing cdc45. siRNA-mediated knockdown was performed to investigate gene functions in HeLa cells overexpressing cdc45. RESULTS We found that high level of cdc45 from different species (human, mammal, ascidian, and Drosophila) inhibited cell cycle in vitro and in vivo. High dose of CDC45 blocks cells entering into S phase. However, we failed to detect DNA damage and cell apoptosis. We identified hspa6 was the most upregulated gene in HeLa cells overexpressing cdc45 via RNA-seq analysis and qRT-PCR validation. Overexpression of Hs-hspa6 inhibited proliferation rate and DNA replication in HeLa cells, mimicking the phenotype of cdc45 overexpression. RNAi against hspa6 partially rescued the cell proliferation defect caused by high dose of CDC45. CONCLUSIONS Our study suggests that high abundance of CDC45 stops cell cycle. Instead of inducing apoptosis, excessive CDC45 prevents cell entering S phase probably due to promoting hspa6 expression.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Sars‐Fang Centre, MoE Key Laboratory of Marine Genetics and BreedingCollege of Marine Life Sciences, Ocean University of ChinaQingdaoChina
| | - Zhiyi Lv
- Sars‐Fang Centre, MoE Key Laboratory of Marine Genetics and BreedingCollege of Marine Life Sciences, Ocean University of ChinaQingdaoChina
- Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Deqing Kong
- Department of BiologyPhilipps UniversityMarburgGermany
| | - Yuping Fan
- Sars‐Fang Centre, MoE Key Laboratory of Marine Genetics and BreedingCollege of Marine Life Sciences, Ocean University of ChinaQingdaoChina
| | - Bo Dong
- Sars‐Fang Centre, MoE Key Laboratory of Marine Genetics and BreedingCollege of Marine Life Sciences, Ocean University of ChinaQingdaoChina
- Institute of Evolution & Marine BiodiversityOcean University of ChinaQingdaoChina
- Laboratory for Marine Biology and BiotechnologyQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
3
|
Botrugno OA, Tonon G. Genomic Instability and Replicative Stress in Multiple Myeloma: The Final Curtain? Cancers (Basel) 2021; 14:cancers14010025. [PMID: 35008191 PMCID: PMC8750813 DOI: 10.3390/cancers14010025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Genomic instability is recognized as a driving force in most cancers as well as in the haematological cancer multiple myeloma and remains among the leading cause of drug resistance. Several evidences suggest that replicative stress exerts a fundamental role in fuelling genomic instability. Notably, cancer cells rely on a single protein, ATR, to cope with the ensuing DNA damage. In this perspective, we provide an overview depicting how replicative stress represents an Achilles heel for multiple myeloma, which could be therapeutically exploited either alone or in combinatorial regimens to preferentially ablate tumor cells. Abstract Multiple Myeloma (MM) is a genetically complex and heterogeneous hematological cancer that remains incurable despite the introduction of novel therapies in the clinic. Sadly, despite efforts spanning several decades, genomic analysis has failed to identify shared genetic aberrations that could be targeted in this disease. Seeking alternative strategies, various efforts have attempted to target and exploit non-oncogene addictions of MM cells, including, for example, proteasome inhibitors. The surprising finding that MM cells present rampant genomic instability has ignited concerted efforts to understand its origin and exploit it for therapeutic purposes. A credible hypothesis, supported by several lines of evidence, suggests that at the root of this phenotype there is intense replicative stress. Here, we review the current understanding of the role of replicative stress in eliciting genomic instability in MM and how MM cells rely on a single protein, Ataxia Telangiectasia-mutated and Rad3-related protein, ATR, to control and survive the ensuing, potentially fatal DNA damage. From this perspective, replicative stress per se represents not only an opportunity for MM cells to increase their evolutionary pool by increasing their genomic heterogeneity, but also a vulnerability that could be leveraged for therapeutic purposes to selectively target MM tumor cells.
Collapse
Affiliation(s)
- Oronza A. Botrugno
- Functional Genomics of Cancer Unit, Experimental Oncology Division, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Correspondence: (O.A.B.); (G.T.); Tel.: +39-02-2643-6661 (O.A.B.); +39-02-2643-5624 (G.T.); Fax: +39-02-2643-6352 (O.A.B. & G.T.)
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Experimental Oncology Division, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Correspondence: (O.A.B.); (G.T.); Tel.: +39-02-2643-6661 (O.A.B.); +39-02-2643-5624 (G.T.); Fax: +39-02-2643-6352 (O.A.B. & G.T.)
| |
Collapse
|
4
|
Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: a multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. J Hematol Oncol 2021; 14:121. [PMID: 34372899 PMCID: PMC8351444 DOI: 10.1186/s13045-021-01111-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022] Open
Abstract
MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and hematopoiesis. Due to the multi-functionality of MYC, its expression is regulated at multiple levels. Deregulation of this oncogene can give rise to a variety of cancers. In this review, MYC regulation and the mechanisms by which MYC adjusts cellular functions and its implication in hematologic malignancies are summarized. Further, we also discuss potential inhibitors of MYC that could be beneficial for treating hematologic malignancies.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Zhang L, Li C, Su X. Emerging impact of the long noncoding RNA MIR22HG on proliferation and apoptosis in multiple human cancers. J Exp Clin Cancer Res 2020; 39:271. [PMID: 33267888 PMCID: PMC7712612 DOI: 10.1186/s13046-020-01784-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
An increasing number of studies have shown that long noncoding RNAs (lncRNAs) play important roles in diverse cellular processes, including proliferation, apoptosis, migration, invasion, chromatin remodeling, metabolism and immune escape. Clinically, the expression of MIR22HG is increased in many human tumors (colorectal cancer, gastric cancer, hepatocellular carcinoma, lung cancer, and thyroid carcinoma), while in others (esophageal adenocarcinoma and glioblastoma), it is significantly decreased. Moreover, MIR22HG has been reported to function as a competitive endogenous RNA (ceRNA), be involved in signaling pathways, interact with proteins and interplay with miRNAs as a host gene to participate in tumorigenesis and tumor progression. In this review, we describe the biological functions of MIR22HG, reveal its underlying mechanisms for cancer regulation, and highlight the potential role of MIR22HG as a novel cancer prognostic biomarker and therapeutic target that can increase the efficacy of immunotherapy and targeted therapy for cancer treatment.
Collapse
Affiliation(s)
- Le Zhang
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, 1 Tong Dao Street, Huimin District, Inner Mongolia, 010050, Hohhot, China
| | - Cuixia Li
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, 1 Tong Dao Street, Huimin District, Inner Mongolia, 010050, Hohhot, China
| | - Xiulan Su
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, 1 Tong Dao Street, Huimin District, Inner Mongolia, 010050, Hohhot, China.
| |
Collapse
|
6
|
Primo LMF, Teixeira LK. DNA replication stress: oncogenes in the spotlight. Genet Mol Biol 2019; 43:e20190138. [PMID: 31930281 PMCID: PMC7197996 DOI: 10.1590/1678-4685gmb-2019-0138] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/09/2019] [Indexed: 01/21/2023] Open
Abstract
Precise replication of genetic material is essential to maintain genome stability. DNA replication is a tightly regulated process that ensues faithful copies of DNA molecules to daughter cells during each cell cycle. Perturbation of DNA replication may compromise the transmission of genetic information, leading to DNA damage, mutations, and chromosomal rearrangements. DNA replication stress, also referred to as DNA replicative stress, is defined as the slowing or stalling of replication fork progression during DNA synthesis as a result of different insults. Oncogene activation, one hallmark of cancer, is able to disturb numerous cellular processes, including DNA replication. In fact, extensive work has indicated that oncogene-induced replication stress is an important source of genomic instability in human carcinogenesis. In this review, we focus on main oncogenes that induce DNA replication stress, such as RAS, MYC, Cyclin E, MDM2, and BCL-2 among others, and the molecular mechanisms by which these oncogenes interfere with normal DNA replication and promote genomic instability.
Collapse
Affiliation(s)
- Luiza M. F. Primo
- Group of Cell Cycle Control, Program of Immunology and Tumor
Biology. Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ,
Brazil
| | - Leonardo K. Teixeira
- Group of Cell Cycle Control, Program of Immunology and Tumor
Biology. Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ,
Brazil
| |
Collapse
|
7
|
Abstract
Precise replication of genetic material is essential to maintain genome stability. DNA replication is a tightly regulated process that ensues faithful copies of DNA molecules to daughter cells during each cell cycle. Perturbation of DNA replication may compromise the transmission of genetic information, leading to DNA damage, mutations, and chromosomal rearrangements. DNA replication stress, also referred to as DNA replicative stress, is defined as the slowing or stalling of replication fork progression during DNA synthesis as a result of different insults. Oncogene activation, one hallmark of cancer, is able to disturb numerous cellular processes, including DNA replication. In fact, extensive work has indicated that oncogene-induced replication stress is an important source of genomic instability in human carcinogenesis. In this review, we focus on main oncogenes that induce DNA replication stress, such as RAS, MYC, Cyclin E, MDM2, and BCL-2 among others, and the molecular mechanisms by which these oncogenes interfere with normal DNA replication and promote genomic instability.
Collapse
Affiliation(s)
- Luiza M F Primo
- Group of Cell Cycle Control, Program of Immunology and Tumor Biology. Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Leonardo K Teixeira
- Group of Cell Cycle Control, Program of Immunology and Tumor Biology. Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
8
|
Dimitrakopoulos C, Hindupur SK, Häfliger L, Behr J, Montazeri H, Hall MN, Beerenwinkel N. Network-based integration of multi-omics data for prioritizing cancer genes. Bioinformatics 2019; 34:2441-2448. [PMID: 29547932 PMCID: PMC6041755 DOI: 10.1093/bioinformatics/bty148] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
Motivation Several molecular events are known to be cancer-related, including genomic aberrations, hypermethylation of gene promoter regions and differential expression of microRNAs. These aberration events are very heterogeneous across tumors and it is poorly understood how they affect the molecular makeup of the cell, including the transcriptome and proteome. Protein interaction networks can help decode the functional relationship between aberration events and changes in gene and protein expression. Results We developed NetICS (Network-based Integration of Multi-omics Data), a new graph diffusion-based method for prioritizing cancer genes by integrating diverse molecular data types on a directed functional interaction network. NetICS prioritizes genes by their mediator effect, defined as the proximity of the gene to upstream aberration events and to downstream differentially expressed genes and proteins in an interaction network. Genes are prioritized for individual samples separately and integrated using a robust rank aggregation technique. NetICS provides a comprehensive computational framework that can aid in explaining the heterogeneity of aberration events by their functional convergence to common differentially expressed genes and proteins. We demonstrate NetICS’ competitive performance in predicting known cancer genes and in generating robust gene lists using TCGA data from five cancer types. Availability and implementation NetICS is available at https://github.com/cbg-ethz/netics. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Christos Dimitrakopoulos
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | | | - Luca Häfliger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jonas Behr
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Hesam Montazeri
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | | | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
9
|
Zhang Y, Li Z, Hao Q, Tan W, Sun J, Li J, Chen CW, Li Z, Meng Y, Zhou Y, Han Z, Pei H, DePamphilis ML, Zhu W. The Cdk2-c-Myc-miR-571 Axis Regulates DNA Replication and Genomic Stability by Targeting Geminin. Cancer Res 2019; 79:4896-4910. [PMID: 31431461 DOI: 10.1158/0008-5472.can-19-0020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/02/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023]
Abstract
DNA rereplication leads to genomic instability and has been implicated in the pathology of a variety of human cancers. Eukaryotic DNA replication is tightly controlled to ensure it occurs only once during each cell cycle. Geminin is a critical component of this control, it prevents DNA rereplication from occurring during S, G2, and early M phases by preventing MCM helicases from forming prereplication complexes. Geminin is targeted for degradation by the anaphase-promoting complex (APC/C) from anaphase through G1-phase, however, accumulating evidence indicates that Geminin is downregulated in late S-phase due to an unknown mechanism. Here, we used a high-throughput screen to identify miRNAs that can induce excess DNA replication and found that miR-571 could reduce the protein level of Geminin in late S-phase independent of the APC/C. Furthermore, miR-571 regulated efficient DNA replication and S-phase cell-cycle progression. Strikingly, c-Myc suppressed miR-571 expression by binding directly to the miR-571 promoter. At the beginning of S-phase, Cdk2 phosphorylated c-Myc at Serine 62, promoting its association with the miR-571 promoter region. Collectively, we identify miR-571 as the first miRNA that prevents aberrant DNA replication and the Cdk2-c-Myc-miR-571 axis as a new pathway for regulating DNA replication, cell cycle, and genomic stability in cancer cells. SIGNIFICANCE: These findings identify a novel regulatory mechanism that is critical for maintaining genome integrity by regulating DNA replication and cell-cycle progression.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Qiang Hao
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Wei Tan
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Jing Sun
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Jing Li
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Chi-Wei Chen
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Zongzhu Li
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Yunxiao Meng
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Yuan Zhou
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Zhiyong Han
- Department of Medical Sciences, Seton Hall-Hackensack Meridian School of Medicine, South Orange, New Jersey
| | - Huadong Pei
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Melvin L DePamphilis
- National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C.
| |
Collapse
|
10
|
Wang Y, Sun B, Zhang Q, Dong H, Zhang J. p300 Acetylates JHDM1A to inhibit osteosarcoma carcinogenesis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2891-2899. [PMID: 31307234 DOI: 10.1080/21691401.2019.1638790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yongkun Wang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baozhen Sun
- Department of Hepatopancreatobiliary Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qiao Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hang Dong
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingzhe Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Brägelmann J, Böhm S, Guthrie MR, Mollaoglu G, Oliver TG, Sos ML. Family matters: How MYC family oncogenes impact small cell lung cancer. Cell Cycle 2017; 16:1489-1498. [PMID: 28737478 DOI: 10.1080/15384101.2017.1339849] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Small cell lung cancer (SCLC) is one of the most deadly cancers and currently lacks effective targeted treatment options. Recent advances in the molecular characterization of SCLC has provided novel insight into the biology of this disease and raises hope for a paradigm shift in the treatment of SCLC. We and others have identified activation of MYC as a driver of susceptibility to Aurora kinase inhibition in SCLC cells and tumors that translates into a therapeutic option for the targeted treatment of MYC-driven SCLC. While MYC shares major features with its paralogs MYCN and MYCL, the sensitivity to Aurora kinase inhibitors is unique for MYC-driven SCLC. In this review, we will compare the distinct molecular features of the 3 MYC family members and address the potential implications for targeted therapy of SCLC.
Collapse
Affiliation(s)
- Johannes Brägelmann
- a Molecular Pathology, Institute of Pathology, University of Cologne , Cologne , Germany.,b Department of Translational Genomics , Medical Faculty, University of Cologne , Cologne , Germany
| | - Stefanie Böhm
- a Molecular Pathology, Institute of Pathology, University of Cologne , Cologne , Germany.,b Department of Translational Genomics , Medical Faculty, University of Cologne , Cologne , Germany
| | - Matthew R Guthrie
- c Department of Oncological Sciences , University of Utah, Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Gurkan Mollaoglu
- c Department of Oncological Sciences , University of Utah, Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Trudy G Oliver
- c Department of Oncological Sciences , University of Utah, Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Martin L Sos
- a Molecular Pathology, Institute of Pathology, University of Cologne , Cologne , Germany.,b Department of Translational Genomics , Medical Faculty, University of Cologne , Cologne , Germany.,d Center for Molecular Medicine Cologne , University of Cologne , Cologne , Germany
| |
Collapse
|
12
|
Ravi D, Beheshti A, Abermil N, Passero F, Sharma J, Coyle M, Kritharis A, Kandela I, Hlatky L, Sitkovsky MV, Mazar A, Gartenhaus RB, Evens AM. Proteasomal Inhibition by Ixazomib Induces CHK1 and MYC-Dependent Cell Death in T-cell and Hodgkin Lymphoma. Cancer Res 2016; 76:3319-31. [PMID: 26988986 DOI: 10.1158/0008-5472.can-15-2477] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
Abstract
Proteasome-regulated NF-κB has been shown to be important for cell survival in T-cell lymphoma and Hodgkin lymphoma models. Several new small-molecule proteasome inhibitors are under various stages of active preclinical and clinical development. We completed a comprehensive preclinical examination of the efficacy and associated biologic effects of a second-generation proteasome inhibitor, ixazomib, in T-cell lymphoma and Hodgkin lymphoma cells and in vivo SCID mouse models. We demonstrated that ixazomib induced potent cell death in all cell lines at clinically achievable concentrations. In addition, it significantly inhibited tumor growth and improved survival in T-cell lymphoma and Hodgkin lymphoma human lymphoma xenograft models. Through global transcriptome analyses, proteasomal inhibition showed conserved overlap in downregulation of cell cycle, chromatin modification, and DNA repair processes in ixazomib-sensitive lymphoma cells. The predicted activity for tumor suppressors and oncogenes, the impact on "hallmarks of cancer," and the analysis of key significant genes from global transcriptome analysis for ixazomib strongly favored tumor inhibition via downregulation of MYC and CHK1, its target genes. Furthermore, in ixazomib-treated lymphoma cells, we identified that CHK1 was involved in the regulation of MYC expression through chromatin modification involving histone H3 acetylation via chromatin immunoprecipitation. Finally, using pharmacologic and RNA silencing of CHK1 or the associated MYC-related mechanism, we demonstrated synergistic cell death in combination with antiproteasome therapy. Altogether, ixazomib significantly downregulates MYC and induces potent cell death in T-cell lymphoma and Hodgkin lymphoma, and we identified that combinatorial therapy with anti-CHK1 treatment represents a rational and novel therapeutic approach. Cancer Res; 76(11); 3319-31. ©2016 AACR.
Collapse
Affiliation(s)
- Dashnamoorthy Ravi
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Afshin Beheshti
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Nasséra Abermil
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Frank Passero
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Jaya Sharma
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Michael Coyle
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Athena Kritharis
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Irawati Kandela
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Lynn Hlatky
- Center of Cancer Systems Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - Michail V Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, Massachusetts
| | - Andrew Mazar
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | | | - Andrew M Evens
- Division of Hematology Oncology and Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts.
| |
Collapse
|
13
|
Chen CH, Lin DS, Cheng CW, Lin CJ, Lo YK, Yen CC, Lee AYL, Hsiao CD. Cdc6 cooperates with c-Myc to promote genome instability and epithelial to mesenchymal transition EMT in zebrafish. Oncotarget 2015; 5:6300-11. [PMID: 25051368 PMCID: PMC4171631 DOI: 10.18632/oncotarget.2204] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aberration in DNA replication is a major cause to genome instability that is a hallmark of cancer cells. Cell division cycle 6 (Cdc6) and c-Myc have a critical role in the initiation of DNA replication. However, whether their interaction induces epithelial-mesenchymal transition (EMT) and promotes tumorigenesis in in vivo animal model remains unclear. Since using zebrafish as a cancer model has been restricted by the late onset of tumorigenesis and extreme difficulty in transformation on skin, we tried to establish a novel non-melanoma skin model in zebrafish to study their role in tumorigenesis. A stable transgenic zebrafish was created by using tol2 transposon, in which cdc6 and c-myc were co-overexpressed in epidermis driven by a skin-specific krt4 promoter. Intriguingly, co-overexpression of cdc6 and c-myc in transgenic zebrafish skin triggered tumor-like transformation, apoptosis attenuation, genomic instability, and EMT, hallmarks of malignant tumorigenesis. Our findings and other characteristics of zebrafish, including optical clarity and small molecule treatment, provide the future utility of this model for easy and non-invasive detection and for identification of new anti-cancer drug.
Collapse
Affiliation(s)
- Ching-Hung Chen
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taiwan
| | - Dar-Shong Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan; Mackay Medical College, Taipei, Taiwan
| | - Chieh-Wen Cheng
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Ju Lin
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taiwan
| | - Yu-Kang Lo
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chueh-Chuan Yen
- Division of Hematology & Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; National Yang-Ming University School of Medicine, Taipei, Taiwan; Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chung-Der Hsiao
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taiwan; Center for Nanotechnology, Chung Yuan Christian University, Chung-Li, Taiwan
| |
Collapse
|
14
|
Zhang Y, Chen HX, Zhou SY, Wang SX, Zheng K, Xu DD, Liu YT, Wang XY, Wang X, Yan HZ, Zhang L, Liu QY, Chen WQ, Wang YF. Sp1 and c-Myc modulate drug resistance of leukemia stem cells by regulating survivin expression through the ERK-MSK MAPK signaling pathway. Mol Cancer 2015; 14:56. [PMID: 25890196 PMCID: PMC4357193 DOI: 10.1186/s12943-015-0326-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/23/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is initiated and maintained by a subset of self-renewing leukemia stem cells (LSCs), which contribute to the progression, recurrence and therapeutic resistance of leukemia. However, the mechanisms underlying the maintenance of LSCs drug resistance have not been fully defined. In this study, we attempted to elucidate the mechanisms of LSCs drug resistance. METHODS We performed reverse phase protein arrays to analyze the expression of anti-apoptotic proteins in the LSC-enriched leukemia cell line KG-1a. Immuno-blotting, cell viability and clinical AML samples were evaluated to verify the micro-assay results. The characteristics and transcriptional regulation of survivin were analyzed with the relative luciferase reporter assay, mutant constructs, chromatin immuno-precipitation (ChIP), quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), and western blotting. The levels of Sp1, c-Myc, phospho-extracellular signal-regulated kinase (p-ERK), phospho-mitogen and stress-activated protein kinase (p-MSK) were investigated in paired CD34+ and CD34- AML patient samples. RESULTS Survivin was highly over-expressed in CD34 + CD38- KG-1a cells and paired CD34+ AML patients compared with their differentiated counterparts. Functionally, survivin contributes to the drug resistance of LSCs, and Sp1 and c-Myc concurrently regulate levels of survivin transcription. Clinically, Sp1 and c-Myc were significantly up-regulated and positively correlated with survivin in CD34+ AML patients. Moreover, Sp1 and c-Myc were further activated by the ERK/MSK mitogen-activated protein kinase (MAPK) signaling pathway, modulating survivin levels. CONCLUSION Our findings demonstrated that ERK/MSK/Sp1/c-Myc axis functioned as a critical regulator of survivin expression in LSCs, offering a potential new therapeutic strategy for LSCs therapy.
Collapse
Affiliation(s)
- Yi Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Hai-xuan Chen
- College of Medicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Shu-yan Zhou
- Department of Pathological Physiology, Wan-nan Medical College, 241000, Wuhu, P.R China.
| | - Shao-xiang Wang
- College of Medicine, Shenzhen University, 518020, Shenzhen, P.R China.
| | - Kai Zheng
- College of Medicine, Shenzhen University, 518020, Shenzhen, P.R China.
| | - Dan-dan Xu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Yu-ting Liu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Xiao-yan Wang
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Xiao Wang
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Hai-Zhao Yan
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Li Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Qiu-ying Liu
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Wan-qun Chen
- College of Medicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Yi-fei Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| |
Collapse
|
15
|
Chayka O, D'Acunto CW, Middleton O, Arab M, Sala A. Identification and pharmacological inactivation of the MYCN gene network as a therapeutic strategy for neuroblastic tumor cells. J Biol Chem 2014; 290:2198-212. [PMID: 25477524 PMCID: PMC4303671 DOI: 10.1074/jbc.m114.624056] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The MYC family of transcription factors consists of three well characterized members, c-MYC, L-MYC, and MYCN, deregulated in the majority of human cancers. In neuronal tumors such as neuroblastoma, MYCN is frequently activated by gene amplification, and reducing its expression by RNA interference has been shown to promote growth arrest and apoptosis of tumor cells. From a clinical perspective, RNA interference is not yet a viable option, and small molecule inhibitors of transcription factors are difficult to develop. We therefore planned to identify, at the global level, the genes interacting functionally with MYCN required to promote fitness of tumor cells facing oncogenic stress. To find genes whose inactivation is synthetically lethal to MYCN, we implemented a genome-wide approach in which we carried out a drop-out shRNA screen using a whole genome library that was delivered into isogenic neuroblastoma cell lines expressing or not expressing MYCN. After the screen, we selected for in-depth analysis four shRNAs targeting AHCY, BLM, PKMYT1, and CKS1B. These genes were chosen because they are directly regulated by MYC proteins, associated with poor prognosis of neuroblastoma patients, and inhibited by small molecule compounds. Mechanistically, we found that BLM and PKMYT1 are required to limit oncogenic stress and promote stabilization of the MYCN protein. Cocktails of small molecule inhibitors of CKS1B, AHCY, BLM, and PKMYT1 profoundly affected the growth of all neuroblastoma cell lines but selectively caused death of MYCN-amplified cells. Our findings suggest that drugging the MYCN network is a promising avenue for the treatment of high risk, neuroblastic cancers.
Collapse
Affiliation(s)
- Olesya Chayka
- From the Brunel Institute of Cancer Genetics and Pharmacogenomics, Brunel University London, London UB8 3PH, United Kingdom and the Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Cosimo Walter D'Acunto
- the Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Odette Middleton
- the Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Maryam Arab
- From the Brunel Institute of Cancer Genetics and Pharmacogenomics, Brunel University London, London UB8 3PH, United Kingdom and
| | - Arturo Sala
- From the Brunel Institute of Cancer Genetics and Pharmacogenomics, Brunel University London, London UB8 3PH, United Kingdom and the Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| |
Collapse
|
16
|
Liu HJ, Wang T, Li QM, Guan XY, Xu Q. Knock-down of p300 decreases the proliferation and odontogenic differentiation potentiality of HDPCs. Int Endod J 2014; 48:976-85. [PMID: 25288362 DOI: 10.1111/iej.12392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 10/02/2014] [Indexed: 01/17/2023]
Abstract
AIM To investigate the role of p300 in the regulation of proliferation and odontogenic differentiation of human dental pulp cells (HDPCs). METHODOLOGY The recombinant lentiviral vector pshRNA-copGFP was used to knock-down p300 expression in HDPCs. Protein level of acetylated H3 was detected. The proliferation of HDPCs was measured using the CCK8 assay. The cell cycle and apoptosis were analysed using flow cytometry and TUNEL staining, respectively. The expression levels of Cdc25A, p21(waf1) and the cleaved products of caspase 3 and caspase 7 were determined utilizing real-time quantitative polymerase chain reaction and Western blotting analysis. The alkaline phosphatase (ALP) activity was measured, and the formation of mineralized nodules was assessed using alizarin red staining after the induction of odontogenic differentiation of HDPCs. The expression levels of the odontogenic differentiation markers DMP-1, DSPP and DSP were detected utilizing real-time quantitative polymerase chain reaction and Western blotting analysis. RESULTS After p300 was knocked down in HDPCs, p300 was significantly down-regulated at both the mRNA and protein levels, and histone H3 acetylation was reduced. The proliferation capacity of HDPCs was suppressed in p300 knock-down groups. The cells were arrested in the G0/G1 phase of the cell cycle, and cell apoptosis was triggered. ALP activity, the formation of mineralized nodules and the expression levels of DMP-1, DSPP and DSP were all decreased in p300-knock-down HDPCs undergoing odontogenic differentiation. CONCLUSION Knocking down p300 restrains the proliferation and odontogenic differentiation potentiality of HDPCs.
Collapse
Affiliation(s)
- H J Liu
- Guanghua School of Stomatology & Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - T Wang
- Guanghua School of Stomatology & Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Q M Li
- Guanghua School of Stomatology & Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - X Y Guan
- Guanghua School of Stomatology & Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Q Xu
- Guanghua School of Stomatology & Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Cazzalini O, Sommatis S, Tillhon M, Dutto I, Bachi A, Rapp A, Nardo T, Scovassi AI, Necchi D, Cardoso MC, Stivala LA, Prosperi E. CBP and p300 acetylate PCNA to link its degradation with nucleotide excision repair synthesis. Nucleic Acids Res 2014; 42:8433-48. [PMID: 24939902 PMCID: PMC4117764 DOI: 10.1093/nar/gku533] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The proliferating cell nuclear antigen (PCNA) protein serves as a molecular platform recruiting and coordinating the activity of factors involved in multiple deoxyribonucleic acid (DNA) transactions. To avoid dangerous genome instability, it is necessary to prevent excessive retention of PCNA on chromatin. Although PCNA functions during DNA replication appear to be regulated by different post-translational modifications, the mechanism regulating PCNA removal and degradation after nucleotide excision repair (NER) is unknown. Here we report that CREB-binding protein (CBP), and less efficiently p300, acetylated PCNA at lysine (Lys) residues Lys13,14,77 and 80, to promote removal of chromatin-bound PCNA and its degradation during NER. Mutation of these residues resulted in impaired DNA replication and repair, enhanced the sensitivity to ultraviolet radiation, and prevented proteolytic degradation of PCNA after DNA damage. Depletion of both CBP and p300, or failure to load PCNA on DNA in NER deficient cells, prevented PCNA acetylation and degradation, while proteasome inhibition resulted in accumulation of acetylated PCNA. These results define a CBP and p300-dependent mechanism for PCNA acetylation after DNA damage, linking DNA repair synthesis with removal of chromatin-bound PCNA and its degradation, to ensure genome stability.
Collapse
Affiliation(s)
- Ornella Cazzalini
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Sabrina Sommatis
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Micol Tillhon
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - Ilaria Dutto
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - Angela Bachi
- IFOM-FIRC Institute of Molecular Oncology, Milan 20100, Italy
| | - Alexander Rapp
- Technische Universität Darmstadt, Darmstadt 64287, Germany
| | - Tiziana Nardo
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - Daniela Necchi
- Department of Drug Sciences, University of Pavia, Pavia 27100, Italy
| | | | - Lucia A Stivala
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Ennio Prosperi
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| |
Collapse
|
18
|
Abstract
The MYC oncogene is a multifunctional protein that is aberrantly expressed in a significant fraction of tumors from diverse tissue origins. Because of its multifunctional nature, it has been difficult to delineate the exact contributions of MYC's diverse roles to tumorigenesis. Here, we review the normal role of MYC in regulating DNA replication as well as its ability to generate DNA replication stress when overexpressed. Finally, we discuss the possible mechanisms by which replication stress induced by aberrant MYC expression could contribute to genomic instability and cancer.
Collapse
Affiliation(s)
| | - Jean Gautier
- Institute for Cancer Genetics, Columbia University, New York, New York 10032 Department of Genetics and Development, Columbia University, New York, New York 10032
| |
Collapse
|
19
|
Wang SA, Hung CY, Chuang JY, Chang WC, Hsu TI, Hung JJ. Phosphorylation of p300 increases its protein degradation to enhance the lung cancer progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1135-49. [PMID: 24530506 DOI: 10.1016/j.bbamcr.2014.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/21/2014] [Accepted: 02/02/2014] [Indexed: 01/05/2023]
Abstract
p300 is a transcription cofactor for a number of nuclear proteins. Most studies of p300 have focused on the regulation of its function, which primarily includes its role as a transcription co-factor for a number of nuclear proteins. In this study, we found that p300 was highly phosphorylated and its level was decreased during mitosis and tumorigenesis. In vitro and in vivo experiments aimed showed that cyclin-dependent kinase 1 (CDK1) and ERK1/2 phosphorylated p300 on Ser1038 and Ser2039. Mutations of Ser1038 and Ser2039 increased p300 protein stability and levels. Inhibition of p300 degradation by blocking its phosphorylation decreased the proliferation and metastasis activity of lung cancer cells, indicating that p300 acts as a tumor suppressor in lung cancer tumorigenesis. Investigation of the molecular mechanism showed that blocking p300 phosphorylation disrupted chromatin condensation and the increased the acetylation of histone H3. Analysis of cell cycle progression in HA-p300-S2A-expressing cells by flow cytometry showed that the p300 mutants arrested the cells at S-phase or delayed the mitotic entry and exit. The expression of several important oncogenes, MMP-9, vimentin, β-catenin, N-cadherin and c-myc, was negatively regulated by p300. In conclusion, during lung tumorigenesis, a phosphorylation-mediated decrease in p300 level enhanced oncogene expression during interphase and decreased histone H3 acetylation during mitosis, which promoted lung cancer progression.
Collapse
Affiliation(s)
- Shao-An Wang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng-Kung University, Tainan 701, Taiwan
| | - Chia-Yang Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan
| | - Jian-Ying Chuang
- Neural Regenerative Medicine, College of Medical Sciences and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wen-Chang Chang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng-Kung University, Tainan 701, Taiwan; Department of Pharmacology, National Cheng-Kung University, Tainan 701, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan; Center for Infectious Disease and Signal Transduction, National Cheng-Kung University, Tainan 701, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsung-I Hsu
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng-Kung University, Tainan 701, Taiwan; Center for Infectious Disease and Signal Transduction, National Cheng-Kung University, Tainan 701, Taiwan
| | - Jan-Jong Hung
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng-Kung University, Tainan 701, Taiwan; Department of Pharmacology, National Cheng-Kung University, Tainan 701, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan; Center for Infectious Disease and Signal Transduction, National Cheng-Kung University, Tainan 701, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
20
|
Leikam C, Hufnagel A, Walz S, Kneitz S, Fekete A, Müller MJ, Eilers M, Schartl M, Meierjohann S. Cystathionase mediates senescence evasion in melanocytes and melanoma cells. Oncogene 2014; 33:771-82. [PMID: 23353821 DOI: 10.1038/onc.2012.641] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 11/28/2012] [Accepted: 12/07/2012] [Indexed: 01/05/2023]
Abstract
The development of malignant melanoma is a highly complex process, which is still poorly understood. A majority of human melanomas are found to express a few oncogenic proteins, such as mutant RAS and BRAF variants. However, these oncogenes are also found in nevi, and it is now a well-accepted fact that their expression alone leads to senescence. This renders the understanding of senescence escape mechanisms an important point to understand tumor development. Here, we approached the question of senescence evasion by expressing the transcription factor v-myc myelocytomatosis viral oncogene homolog (c-MYC), which is known to act synergistically with many oncogenes, in melanocytes. We observed that MYC drives the evasion of reactive-oxygen stress-induced melanocyte senescence, caused by activated receptor tyrosine kinase signaling. Conversely, MIZ1, the growth suppressing interaction partner of MYC, is involved in mediating melanocyte senescence. Both, MYC overexpression and Miz1 knockdown led to a strong reduction of endogenous reactive-oxygen species (ROS), DNA damage and senescence. We identified the cystathionase (CTH) gene product as mediator of the ROS-related MYC and MIZ1 effects. Blocking CTH enzymatic activity in MYC-overexpressing and Miz1 knockdown cells increased intracellular stress and senescence. Importantly, pharmacological inhibition of CTH in human melanoma cells also reconstituted senescence in the majority of cell lines, and CTH knockdown reduced tumorigenic effects such as proliferation, H2O2 resistance and soft agar growth. Thus, we identified CTH as new MYC target gene with an important function in senescence evasion.
Collapse
Affiliation(s)
- C Leikam
- Department of Physiological Chemistry I, Biocenter, University of Wurzburg, Wurzburg, Germany
| | - A Hufnagel
- Department of Physiological Chemistry I, Biocenter, University of Wurzburg, Wurzburg, Germany
| | - S Walz
- Department of Biochemistry and Molecular Biology, Biocenter, University of Wurzburg, Wurzburg, Germany
| | - S Kneitz
- Department of Physiological Chemistry I, Biocenter, University of Wurzburg, Wurzburg, Germany
| | - A Fekete
- Department of Pharmaceutical Biology, Julius-von-Sachs Institute for Biological Sciences, University of Wurzburg, Wurzburg, Germany
| | - M J Müller
- Department of Pharmaceutical Biology, Julius-von-Sachs Institute for Biological Sciences, University of Wurzburg, Wurzburg, Germany
| | - M Eilers
- Department of Biochemistry and Molecular Biology, Biocenter, University of Wurzburg, Wurzburg, Germany
| | - M Schartl
- Department of Physiological Chemistry I, Biocenter, University of Wurzburg, Wurzburg, Germany
| | - S Meierjohann
- Department of Physiological Chemistry I, Biocenter, University of Wurzburg, Wurzburg, Germany
| |
Collapse
|
21
|
Urbanek-Olejnik K, Liszewska M, Winczura A, Kostka G. Changes of c-Myc and DNMT1 mRNA and protein levels in the rat livers induced by dibutyl phthalate treatment. Toxicol Ind Health 2013; 32:801-8. [DOI: 10.1177/0748233713512363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We investigated the relationship between dibutyl phthalate (DBP)-induced hypomethylation of the c-Myc promoter region (as evident in our early study) and the expression of c-Myc and DNMT1 genes (at messenger RNA (mRNA) and protein level) in the rat liver. Male Wistar rats received DBP in 1, 3, or 14 daily doses of 1800 mg kg−1 body weight. Levels of DNMT1, c-Myc mRNA, and proteins were detected using real-time polymerase chain reaction and Western blot analysis, respectively. Our findings indicate that DBP caused an increase in mRNA levels of c-Myc at all time points. The results showed that protein levels of c-Myc in rat liver also increased significantly by DBP treatment, which were more pronounced at last time point (after 14 doses). Furthermore, overexpression of DNMT1gene have been found after one dose of DBP, which was confirmed at the protein level by Western blot analysis. Reduced levels of DNMT1mRNA and proteins (3 and 14 doses) were coordinated with depletion DNA synthesis (reported previously). Based on our previous results and those presented here, the following conclusion could be drawn: (1) DBP exerted biological activity through epigenetic modulation of c-Myc gene expression; (2) it seems possible that DBP-induced active demethylation of c-Myc gene through mechanism(s) linked to generation of reactive oxygen species by activated c-Myc; and (3) control of DNA replication was not directly dependent on c-Myc transcriptional activity and we attribute this finding to DNMT1gene expression which was tightly coordinated with DNA synthesis.
Collapse
Affiliation(s)
- Katarzyna Urbanek-Olejnik
- Department of Toxicology and Risk Assessment, National Institute of Public Health-National Institute of Hygiene, Warsaw, Poland
| | - Monika Liszewska
- Department of Toxicology and Risk Assessment, National Institute of Public Health-National Institute of Hygiene, Warsaw, Poland
| | - Alicja Winczura
- Department of Molecular Biology, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Grażyna Kostka
- Department of Toxicology and Risk Assessment, National Institute of Public Health-National Institute of Hygiene, Warsaw, Poland
| |
Collapse
|
22
|
Valero ML, Cimas FJ, Arias L, Melgar-Rojas P, García E, Callejas-Valera JL, García-Cano J, Serrano-Oviedo L, Ángel de la Cruz-Morcillo M, Sánchez-Pérez I, Sánchez-Prieto R. E1a promotes c-Myc-dependent replicative stress: implications in glioblastoma radiosensitization. Cell Cycle 2013; 13:52-61. [PMID: 24196438 PMCID: PMC3925735 DOI: 10.4161/cc.26754] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/08/2013] [Accepted: 10/08/2013] [Indexed: 12/27/2022] Open
Abstract
The E1a gene from adenovirus is known to be a potent inducer of chemo/radiosensitivity in a wide range of tumors. However, the molecular bases of its radiosensitizer properties are still poorly understood. In an attempt to study this effect, U87MG cells, derived from a radio-resistant tumor as glioblastoma, where infected with lentivirus carrying E1a gene developing an acute sensitivity to ionizing radiation. The induction of radiosensitivity correlated with a marked G 2/M phase accumulation and a potent apoptotic response. Our findings demonstrate that c-Myc plays a pivotal role in E1a-associated radiosensitivity through the induction of a replicative stress situation, as our data support by genetic approaches, based in interference and overexpression in U87MG cells. In fact, we present evidence showing that Chk1 is a novel transcriptional target of E1a gene through the effect exerted by this adenoviral protein onto c-Myc. Moreover, c-Myc upregulation also explains the marked phosphorylation of H2AX associated to E1a expression in the absence of DNA damage. Indeed, all these observations were applicable to other experimental models, such as T98G, LN-405 and A172, rendering the same pattern in terms of radiosensitivity, cell cycle distribution, upregulation of Chk1, c-Myc, and phosphorylation pattern of H2AX. In summary, our data propose a novel mechanism to explain how E1a mediates radiosensitivity through the signaling axis E1a→c-Myc→ replicative stress situation. This novel mechanism of E1a-mediated radiosensitivity could be the key to open new possibilities in the current therapy of glioblastoma.
Collapse
Affiliation(s)
- María Llanos Valero
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Francisco Jose Cimas
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Laura Arias
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Pedro Melgar-Rojas
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Elena García
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Juan Luis Callejas-Valera
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Jesús García-Cano
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Leticia Serrano-Oviedo
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Miguel Ángel de la Cruz-Morcillo
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Isabel Sánchez-Pérez
- Department of Biochemistry; School of Medicine;Biomedical Research Institute of Madrid CSIC/UAM; Madrid, Spain
| | - Ricardo Sánchez-Prieto
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| |
Collapse
|
23
|
Adenovirus E1A oncogene induces rereplication of cellular DNA and alters DNA replication dynamics. J Virol 2013; 87:8767-78. [PMID: 23740993 DOI: 10.1128/jvi.00879-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The oncogenic property of the adenovirus (Ad) transforming E1A protein is linked to its capacity to induce cellular DNA synthesis which occurs as a result of its interaction with several host proteins, including pRb and p300/CBP. While the proteins that contribute to the forced induction of cellular DNA synthesis have been intensively studied, the nature of the cellular DNA replication that is induced by E1A in quiescent cells is not well understood. Here we show that E1A expression in quiescent cells leads to massive cellular DNA rereplication in late S phase. Using a single-molecule DNA fiber assay, we studied the cellular DNA replication dynamics in E1A-expressing cells. Our studies show that the DNA replication pattern is dramatically altered in E1A-expressing cells, with increased replicon length, fork velocity, and interorigin distance. The interorigin distance increased by about 3-fold, suggesting that fewer DNA replication origins are used in E1A-expressing cells. These aberrant replication events led to replication stress, as evidenced by the activation of the DNA damage response. In earlier studies, we showed that E1A induces c-Myc as a result of E1A binding to p300. Using an antisense c-Myc to block c-Myc expression, our results indicate that induction of c-Myc in E1A-expressing cells contributes to the induction of host DNA replication. Together, our results suggest that the E1A oncogene-induced cellular DNA replication stress is due to dramatically altered cellular replication events and that E1A-induced c-Myc may contribute to these events.
Collapse
|
24
|
Srinivasan SV, Dominguez-Sola D, Wang LC, Hyrien O, Gautier J. Cdc45 is a critical effector of myc-dependent DNA replication stress. Cell Rep 2013; 3:1629-39. [PMID: 23643534 DOI: 10.1016/j.celrep.2013.04.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 01/02/2013] [Accepted: 04/03/2013] [Indexed: 01/31/2023] Open
Abstract
c-Myc oncogenic activity is thought to be mediated in part by its ability to generate DNA replication stress and subsequent genomic instability when deregulated. Previous studies have demonstrated a nontranscriptional role for c-Myc in regulating DNA replication. Here, we analyze the mechanisms by which c-Myc deregulation generates DNA replication stress. We find that overexpression of c-Myc alters the spatiotemporal program of replication initiation by increasing the density of early-replicating origins. We further show that c-Myc deregulation results in elevated replication-fork stalling or collapse and subsequent DNA damage. Notably, these phenotypes are independent of RNA transcription. Finally, we demonstrate that overexpression of Cdc45 recapitulates all c-Myc-induced replication and damage phenotypes and that Cdc45 and GINS function downstream of Myc.
Collapse
|
25
|
Tillhon M, Cazzalini O, Nardo T, Necchi D, Sommatis S, Stivala LA, Scovassi AI, Prosperi E. p300/CBP acetyl transferases interact with and acetylate the nucleotide excision repair factor XPG. DNA Repair (Amst) 2012; 11:844-52. [PMID: 22954786 DOI: 10.1016/j.dnarep.2012.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/03/2012] [Accepted: 08/03/2012] [Indexed: 01/01/2023]
Abstract
Nucleotide excision repair (NER) is an important DNA repair mechanism through which cells remove bulky DNA lesions. Following DNA damage, the histone acetyltransferase (HAT) p300 (also referred to as lysine acetyltransferase or KAT) is known to associate with proliferating cell nuclear antigen (PCNA), a master regulator of DNA replication and repair processes. This interaction, which results in HAT inhibition, may be dissociated by the cell cycle inhibitor p21(CDKN1A), thereby restoring p300 activity; however, the role of this protein interplay is still unclear. Here, we report that silencing p300 or its homolog CREB-binding protein (CBP) by RNA interference (RNAi) significantly reduces DNA repair synthesis in human fibroblasts. In addition, we determined whether p300 and CBP may associate with and acetylate specific NER factors such as XPG, the 3'-endonuclease that is involved in the incision/excision step and is known to interact with PCNA. Our results show that p300 and CBP interact with XPG, which has been found to be acetylated in vivo. XPG is acetylated by p300 in vitro, and this reaction is inhibited by PCNA. Knocking down both p300/CBP by RNAi or by chemical inhibition with curcumin greatly reduced XPG acetylation, and a concomitant accumulation of the protein at DNA damage sites was observed. The ability of p21 to bind PCNA was found to regulate the interaction between p300 and XPG, and an abnormal accumulation of XPG at DNA damage sites was also found in p21(-/-) fibroblasts. These results indicate an additional function of p300/CBP in NER through the acetylation of XPG protein in a PCNA-p21 dependent manner.
Collapse
Affiliation(s)
- Micol Tillhon
- Istituto di Genetica Molecolare (IGM) del CNR, Pavia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Takezawa K, Okamoto I, Tsukioka S, Uchida J, Kiniwa M, Fukuoka M, Nakagawa K. Identification of thymidylate synthase as a potential therapeutic target for lung cancer. Br J Cancer 2010; 103:354-61. [PMID: 20628382 PMCID: PMC2920030 DOI: 10.1038/sj.bjc.6605793] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background: Thymidylate synthase (TS), a key enzyme in the de novo synthesis of thymidine, is an important chemotherapeutic target for malignant tumours including lung cancer. Although inhibition of TS has an antiproliferative effect in cancer cells, the precise mechanism of this effect has remained unclear. Methods: We examined the effects of TS inhibition with an RNA interference-based approach. The effect of TS depletion on the growth of lung cancer cells was examined using colorimetric assay and flow cytometry. Results: Measurement of the enzymatic activity of TS in 30 human lung cancer cell lines revealed that such activity differs among tumour histotypes. Almost complete elimination of TS activity by RNA interference resulted in inhibition of cell proliferation in all tested cell lines, suggestive of a pivotal role for TS in cell proliferation independent of the original level of enzyme activity. The antiproliferative effect of TS depletion was accompanied by arrest of cells in S phase of the cell cycle and the induction of caspase-dependent apoptosis as well as by changes in the expression levels of cyclin E and c-Myc. Moreover, TS depletion induced downregulation of the antiapoptotic protein X-linked inhibitor of apoptosis (XIAP), and it seemed to activate the mitochondrial pathway of apoptosis. Conclusion: Our data provide insight into the biological relevance of TS as well as a basis for clinical development of TS-targeted therapy for lung cancer.
Collapse
Affiliation(s)
- K Takezawa
- Department of Medical Oncology, Kinki University School of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Cha H, Lowe JM, Li H, Lee JS, Belova GI, Bulavin DV, Fornace AJ. Wip1 directly dephosphorylates gamma-H2AX and attenuates the DNA damage response. Cancer Res 2010; 70:4112-22. [PMID: 20460517 DOI: 10.1158/0008-5472.can-09-4244] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The integrity of DNA is constantly challenged throughout the life of a cell by both endogenous and exogenous stresses. A well-organized rapid damage response and proficient DNA repair, therefore, become critically important for maintaining genomic stability and cell survival. When DNA is damaged, the DNA damage response (DDR) can be initiated by alterations in chromosomal structure and histone modifications, such as the phosphorylation of the histone H2AX (the phosphorylated form is referred to as gamma-H2AX). gamma-H2AX plays a crucial role in recruiting DDR factors to damage sites for accurate DNA repair. On repair completion, gamma-H2AX must then be reverted to H2AX by dephosphorylation for attenuation of the DDR. Here, we report that the wild-type p53-induced phosphatase 1 (Wip1) phosphatase, which is often overexpressed in a variety of tumors, effectively dephosphorylates gamma-H2AX in vitro and in vivo. Ectopic expression of Wip1 significantly reduces the level of gamma-H2AX after ionizing as well as UV radiation. Forced premature dephosphorylation of gamma-H2AX by Wip1 disrupts recruitment of important DNA repair factors to damaged sites and delays DNA damage repair. Additionally, deletion of Wip1 enhances gamma-H2AX levels in cells undergoing constitutive oncogenic stress. Taken together, our studies show that Wip1 is an important mammalian phosphatase for gamma-H2AX and shows an additional mechanism for Wip1 in the tumor surveillance network.
Collapse
Affiliation(s)
- Hyukjin Cha
- Department of Biochemistry, Georgetown University, Washington, District of Columbia 20057-1468, USA.
| | | | | | | | | | | | | |
Collapse
|