1
|
Park J, Lee DH. Loss of protein phosphatase 4 inhibitory protein leads to genomic instability and heightens vulnerability to replication stress. Biochim Biophys Acta Gen Subj 2025; 1869:130797. [PMID: 40157551 DOI: 10.1016/j.bbagen.2025.130797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Protein phosphatase 4 inhibitory protein (PP4IP) has recently emerged as a key player in cellular processes, particularly in DNA double-strand break repair and telomere maintenance, although research on its functions remains limited. To further investigate the cellular pathways involving PP4IP, we conducted transcriptomic analysis via RNA sequencing in PP4IP-knockout cells and observed an upregulation of p21 expression. This upregulation was linked to an increased population of p21-positive G1-phase cells in the absence of PP4IP. Prior studies have suggested that unresolved under-replicated DNA in mother cells, transmitted to daughter cells, can trigger a quiescent G1 phase characterized by p21 expression and the formation of p53-binding protein 1 (53BP1) nuclear bodies. Consistent with this, we found a higher proportion of 53BP1 nuclear bodies-positive G1 cells in PP4IP-knockout cells compared to controls. Additionally, PP4IP-deficient cells displayed an increased occurrence of anaphase bridges-indicative of incomplete DNA replication-without a corresponding increase in lagging chromosomes. Furthermore, PP4IP-knockout cells exhibited a heightened susceptibility to replication stress, as evidenced by an elevated frequency of replication stress-induced chromatid breaks and increased sensitivity to such stress. Collectively, these results suggest that PP4IP plays a critical role in safeguarding cells from replication stress and ensuring genomic stability.
Collapse
Affiliation(s)
- Jaehong Park
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea.
| | - Dong-Hyun Lee
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea; Research Center of Ecomimetics, Chonnam National University, Gwangju, Republic of Korea; Institute of Sustainable Ecological Environment, Chonnam National Univesity, Gwangju, Republic of Korea.
| |
Collapse
|
2
|
Ceccaldi R, Cejka P. Mechanisms and regulation of DNA end resection in the maintenance of genome stability. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00841-4. [PMID: 40133633 DOI: 10.1038/s41580-025-00841-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 03/27/2025]
Abstract
DNA end resection is a crucial early step in most DNA double-strand break (DSB) repair pathways. Resection involves the nucleolytic degradation of 5' ends at DSB sites to generate 3' single-stranded DNA overhangs. The first, short-range resection step is catalysed by the nuclease MRE11, acting as part of the MRE11-RAD50-NBS1 complex. Subsequent long-range resection is catalysed by the nucleases EXO1 and/or DNA2. Resected DNA is necessary for homology search and the priming of DNA synthesis in homologous recombination. DNA overhangs may also mediate DNA annealing in the microhomology-mediated end-joining and single-strand annealing pathways, and activate the DNA damage response. By contrast, DNA end resection inhibits DSB repair by non-homologous end-joining. In this Review, we discuss the importance of DNA end resection in various DSB repair pathways, the molecular mechanisms of end resection and its regulation, focusing on phosphorylation and other post-translational modifications that control resection throughout the cell cycle and in response to DNA damage.
Collapse
Affiliation(s)
- Raphael Ceccaldi
- INSERM U830, PSL Research University, Institut Curie, Paris, France.
| | - Petr Cejka
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
3
|
Billing D, Sfeir A. The Role of Microhomology-Mediated End Joining (MMEJ) at Dysfunctional Telomeres. Cold Spring Harb Perspect Biol 2025; 17:a041687. [PMID: 39500624 PMCID: PMC11864110 DOI: 10.1101/cshperspect.a041687] [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] [Indexed: 02/05/2025]
Abstract
DNA double-strand break (DSB) repair pathways are crucial for maintaining genome stability and cell viability. However, these pathways can mistakenly recognize chromosome ends as DNA breaks, leading to adverse outcomes such as telomere fusions and malignant transformation. The shelterin complex protects telomeres from activation of DNA repair pathways by inhibiting nonhomologous end joining (NHEJ), homologous recombination (HR), and microhomology-mediated end joining (MMEJ). The focus of this paper is on MMEJ, an error-prone DSB repair pathway characterized by short insertions and deletions flanked by sequence homology. MMEJ is critical in mediating telomere fusions in cells lacking the shelterin complex and at critically short telomeres. Furthermore, studies suggest that MMEJ is the preferred pathway for repairing intratelomeric DSBs and facilitates escape from telomere crisis. Targeting MMEJ to prevent telomere fusions in hematologic malignancies is of potential therapeutic value.
Collapse
Affiliation(s)
- David Billing
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Agnel Sfeir
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
4
|
Meyer‐Gerards C, Bazzi H. Developmental and tissue-specific roles of mammalian centrosomes. FEBS J 2025; 292:709-726. [PMID: 38935637 PMCID: PMC11839934 DOI: 10.1111/febs.17212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/08/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024]
Abstract
Centrosomes are dominant microtubule organizing centers in animal cells with a pair of centrioles at their core. They template cilia during interphase and help organize the mitotic spindle for a more efficient cell division. Here, we review the roles of centrosomes in the early developing mouse and during organ formation. Mammalian cells respond to centrosome loss-of-function by activating the mitotic surveillance pathway, a timing mechanism that, when a defined mitotic duration is exceeded, leads to p53-dependent cell death in the descendants. Mouse embryos without centrioles are highly susceptible to this pathway and undergo embryonic arrest at mid-gestation. The complete loss of the centriolar core results in earlier and more severe phenotypes than that of other centrosomal proteins. Finally, different developing tissues possess varying thresholds and mount graded responses to the loss of centrioles that go beyond the germ layer of origin.
Collapse
Affiliation(s)
- Charlotte Meyer‐Gerards
- Department of Cell Biology of the Skin, Medical FacultyUniversity of CologneGermany
- Department of Dermatology and Venereology, Medical FacultyUniversity of CologneGermany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging‐associated Diseases (CECAD), Medical FacultyUniversity of CologneGermany
- Graduate School for Biological SciencesUniversity of CologneGermany
- Center for Molecular Medicine Cologne (CMMC), Medical FacultyUniversity of CologneGermany
| | - Hisham Bazzi
- Department of Cell Biology of the Skin, Medical FacultyUniversity of CologneGermany
- Department of Dermatology and Venereology, Medical FacultyUniversity of CologneGermany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging‐associated Diseases (CECAD), Medical FacultyUniversity of CologneGermany
- Center for Molecular Medicine Cologne (CMMC), Medical FacultyUniversity of CologneGermany
- Present address:
Cell & Developmental BiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
5
|
Belal H, Ying Ng EF, Meitinger F. 53BP1-mediated activation of the tumor suppressor p53. Curr Opin Cell Biol 2024; 91:102424. [PMID: 39244835 DOI: 10.1016/j.ceb.2024.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
In recent years, the role of 53BP1 as a cell cycle regulator has come into the spotlight. 53BP1 is best understood for its role in controlling DNA double-strand break repair. However, 53BP1 was initially discovered as an interaction partner of the tumor suppressor p53, which proved to be independent of DNA repair. The importance of this interaction is becoming increasingly clear. 53BP1 responds to mitotic stress, which prolongs mitosis, or to DNA damage and triggers the stabilization of p53 by the deubiquitinase USP28 to stop the proliferation of potentially damaged cells. The ability of 53BP1 to respond to mitotic stress or DNA damage is controlled by cell cycle-specific post-translational modifications and is therefore restricted to specific cell cycle phases. 53BP1-mediated p53 activation is likely involved in tumor suppression and is associated with genetic diseases such as primary microcephaly. This review emphasizes the importance of these mechanisms for the development and maintenance of healthy tissues.
Collapse
Affiliation(s)
- Hazrat Belal
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Esther Feng Ying Ng
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Franz Meitinger
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan.
| |
Collapse
|
6
|
Kilgas S, Swift ML, Chowdhury D. 53BP1-the 'Pandora's box' of genome integrity. DNA Repair (Amst) 2024; 144:103779. [PMID: 39476547 DOI: 10.1016/j.dnarep.2024.103779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
53BP1 has several functions in the maintenance of genome integrity. It functions as a key mediator involved in double-strand break (DSB) repair, which functions to maintain a balance in the repair pathway choices and in preserving genomic stability. While its DSB repair functions are relatively well-characterized, its role in DNA replication and replication fork protection is less understood. In response to replication stress, 53BP1 contributes to fork protection by regulating fork reversal and restart. It helps maintain replication fork stability and speed, with 53BP1 loss leading to defective fork progression and increased sensitivity to replication stress agents. However, 53BP1's precise role in fork protection remains debated, as some studies have not observed protective effects. Therefore, it is critical to determine the role of 53BP1 in replication to better understand when it promotes replication fork protection, and the underlying mechanisms involved. Moreover, 53BP1's function in replication stress extends beyond its activity at active replication forks; it also forms specialized nuclear bodies (NBs) which protect stretches of under-replicated DNA (UR-DNA) transmitted from a previous cell cycle to daughter cells through mitosis. The mechanism of 53BP1 NBs in the coordination of replication and repair events at UR-DNA loci is not fully understood and warrants further investigation. The present review article focuses on elucidating 53BP1's functions in replication stress (RS), its role in replication fork protection, and the significance of 53BP1 NBs in this context to provide a more comprehensive understanding of its less well-established role in DNA replication.
Collapse
Affiliation(s)
- Susan Kilgas
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Michelle L Swift
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Dipanjan Chowdhury
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Joo YK, Ramirez C, Kabeche L. A TRilogy of ATR's Non-Canonical Roles Throughout the Cell Cycle and Its Relation to Cancer. Cancers (Basel) 2024; 16:3536. [PMID: 39456630 PMCID: PMC11506335 DOI: 10.3390/cancers16203536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Ataxia Telangiectasia and Rad3-related protein (ATR) is an apical kinase of the DNA Damage Response (DDR) pathway responsible for detecting and resolving damaged DNA. Because cancer cells depend heavily on the DNA damage checkpoint for their unchecked proliferation and propagation, ATR has gained enormous popularity as a cancer therapy target in recent decades. Yet, ATR inhibitors have not been the silver bullets as anticipated, with clinical trials demonstrating toxicity and mixed efficacy. To investigate whether the toxicity and mixed efficacy of ATR inhibitors arise from their off-target effects related to ATR's multiple roles within and outside the DDR pathway, we have analyzed recently published studies on ATR's non-canonical roles. Recent studies have elucidated that ATR plays a wide role throughout the cell cycle that is separate from its function in the DDR. This includes maintaining nuclear membrane integrity, detecting mechanical forces, and promoting faithful chromosome segregation during mitosis. In this review, we summarize the canonical, DDR-related roles of ATR and also focus on the non-canonical, multifaceted roles of ATR throughout the cell cycle and their clinical relevance. Through this summary, we also address the need for re-assessing clinical strategies targeting ATR as a cancer therapy based on these newly discovered roles for ATR.
Collapse
Affiliation(s)
- Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Carlos Ramirez
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| |
Collapse
|
8
|
Sfeir A, Tijsterman M, McVey M. Microhomology-Mediated End-Joining Chronicles: Tracing the Evolutionary Footprints of Genome Protection. Annu Rev Cell Dev Biol 2024; 40:195-218. [PMID: 38857538 DOI: 10.1146/annurev-cellbio-111822-014426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The fidelity of genetic information is essential for cellular function and viability. DNA double-strand breaks (DSBs) pose a significant threat to genome integrity, necessitating efficient repair mechanisms. While the predominant repair strategies are usually accurate, paradoxically, error-prone pathways also exist. This review explores recent advances and our understanding of microhomology-mediated end joining (MMEJ), an intrinsically mutagenic DSB repair pathway conserved across organisms. Central to MMEJ is the activity of DNA polymerase theta (Polθ), a specialized polymerase that fuels MMEJ mutagenicity. We examine the molecular intricacies underlying MMEJ activity and discuss its function during mitosis, where the activity of Polθ emerges as a last-ditch effort to resolve persistent DSBs, especially when homologous recombination is compromised. We explore the promising therapeutic applications of targeting Polθ in cancer treatment and genome editing. Lastly, we discuss the evolutionary consequences of MMEJ, highlighting its delicate balance between protecting genome integrity and driving genomic diversity.
Collapse
Affiliation(s)
- Agnel Sfeir
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA;
| | - Marcel Tijsterman
- Department of Human Genetics, Leiden University Medical Center; Institute of Biology Leiden, Leiden University, Leiden, The Netherlands;
| | - Mitch McVey
- Department of Biology, Tufts University, Medford, Massachusetts, USA;
| |
Collapse
|
9
|
Dong MZ, Ouyang YC, Gao SC, Gu LJ, Guo JN, Sun SM, Wang ZB, Sun QY. Protein phosphatase 4 maintains the survival of primordial follicles by regulating autophagy in oocytes. Cell Death Dis 2024; 15:658. [PMID: 39245708 PMCID: PMC11381532 DOI: 10.1038/s41419-024-07051-4] [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: 03/23/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
In mammalian ovary, the primordial follicle pool serves as the source of developing follicles and fertilizable ova. To maintain the normal length of female reproductive life, the primordial follicles must have adequate number and be kept in a quiescent state before menopause. However, the molecular mechanisms underlying primordial follicle survival are poorly understood. Here, we provide genetic evidence showing that lacking protein phosphatase 4 (PPP4) in oocytes, a member of PP2A-like subfamily, results in infertility in female mice. A large quantity of primordial follicles has been depleted around the primordial follicle pool formation phase and the ovarian reserve is exhausted at about 7 months old. Further investigation demonstrates that depletion of PPP4 causes the abnormal activation of mTOR, which suppresses autophagy in primordial follicle oocytes. The abnormal primordial follicle oocytes are eventually erased by pregranulosa cells in the manner of lysosome invading. These results show that autophagy prevents primordial follicles over loss and PPP4-mTOR pathway governs autophagy during the primordial follicle formation and dormant period.
Collapse
Affiliation(s)
- Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shi-Cai Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lin-Jian Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia-Ni Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si-Min Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| |
Collapse
|
10
|
Wang J, Sadeghi CA, Le LV, Le Bouteiller M, Frock RL. ATM and 53BP1 regulate alternative end joining-mediated V(D)J recombination. SCIENCE ADVANCES 2024; 10:eadn4682. [PMID: 39083600 PMCID: PMC11290492 DOI: 10.1126/sciadv.adn4682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 06/11/2024] [Indexed: 08/02/2024]
Abstract
G0-G1 phase alternative end joining (A-EJ) is a recently defined mutagenic pathway characterized by resected deletion and translocation joints that are predominantly direct and are distinguished from A-EJ in cycling cells that rely much more on microhomology-mediated end joining (MMEJ). Using chemical and genetic approaches, we systematically evaluate potential A-EJ factors and DNA damage response (DDR) genes to support this mechanism by mapping the repair fates of RAG1/2-initiated double-strand breaks in the context of Igκ locus V-J recombination and chromosome translocation. Our findings highlight a polymerase theta-independent Parp1-XRCC1/LigIII axis as central A-EJ components, supported by 53BP1 in the context of an Ataxia-telangiectasia mutated (ATM)-activated DDR. Mechanistically, we demonstrate varied changes in short-range resection, MMEJ, and translocation, imposed by compromising specific DDR activities, which include polymerase alpha, Ataxia-telangiectasia and Rad3-related (ATR), DNA2, and Mre11. This study advances our understanding of DNA damage repair within the 53BP1 regulatory domain and the RAG1/2 postcleavage complex.
Collapse
Affiliation(s)
- Jinglong Wang
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cheyenne A. Sadeghi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Long V. Le
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marie Le Bouteiller
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
11
|
Perdikopanis N, Giannakakis A, Kavakiotis I, Hatzigeorgiou AG. D-sORF: Accurate Ab Initio Classification of Experimentally Detected Small Open Reading Frames (sORFs) Associated with Translational Machinery. BIOLOGY 2024; 13:563. [PMID: 39194501 DOI: 10.3390/biology13080563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Small open reading frames (sORFs; <300 nucleotides or <100 amino acids) are widespread across all genomes, and an increasing variety of them appear to be translating from non-genic regions. Over the past few decades, peptides produced from sORFs have been identified as functional in various organisms, from bacteria to humans. Despite recent advances in next-generation sequencing and proteomics, accurate annotation and classification of sORFs remain a rate-limiting step toward reliable and high-throughput detection of small proteins from non-genic regions. Additionally, the cost of computational methods utilizing machine learning is lower than that of biological experiments, and they can be employed to detect sORFs, laying the groundwork for biological experiments. We present D-sORF, a machine-learning framework that integrates the statistical nucleotide context and motif information around the start codon to predict coding sORFs. D-sORF scores directly for coding identity and requires only the underlying genomic sequence, without incorporating parameters such as the conservation, which, in the case of sORFs, may increase the dispersion of scores within the significantly less conserved non-genic regions. D-sORF achieves 94.74% precision and 92.37% accuracy for small ORFs (using the 99 nt medium length window). When D-sORF is applied to sORFs associated with ribosomes, the identification of transcripts producing peptides (annotated by the Ensembl IDs) is similar to or superior to experimental methodologies based on ribosome-sequencing (Ribo-Seq) profiling. In parallel, the recognition of putative negative data, such as the intron-containing transcripts that associate with ribosomes, remains remarkably low, indicating that D-sORF could be efficiently applied to filter out false-positive sORFs from Ribo-Seq data because of the non-productive ribosomal binding or noise inherent in these protocols.
Collapse
Affiliation(s)
- Nikos Perdikopanis
- Department of Electrical and Computer Engineering, University of Thessaly, 38221 Volos, Greece
- Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, 15784 Athens, Greece
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 38221 Volos, Greece
| | - Antonis Giannakakis
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kavakiotis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 38221 Volos, Greece
| | - Artemis G Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 38221 Volos, Greece
- Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
12
|
Cokelaere C, Dok R, Cortesi EE, Zhao P, Sablina A, Nuyts S, Derua R, Janssens V. TIPRL1 and its ATM-dependent phosphorylation promote radiotherapy resistance in head and neck cancer. Cell Oncol (Dordr) 2024; 47:793-818. [PMID: 37971644 DOI: 10.1007/s13402-023-00895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE TIPRL1 (target of rapamycin signaling pathway regulator-like 1) is a known interactor and inhibitor of protein phosphatases PP2A, PP4 and PP6 - all pleiotropic modulators of the DNA Damage Response (DDR). Here, we investigated the role of TIPRL1 in the radiotherapy (RT) response of Head and Neck Squamous Cell Carcinoma (HNSCC). METHODS TIPRL1 mRNA (cBioportal) and protein expression (immunohistochemistry) in HNSCC samples were linked with clinical patient data. TIPRL1-depleted HNSCC cells were generated by CRISPR/Cas9 editing, and effects on colony growth, micronuclei formation (microscopy), cell cycle (flow cytometry), DDR signaling (immunoblots) and proteome (mass spectrometry) following RT were assessed. Mass spectrometry was used for TIPRL1 phosphorylation and interactomics analysis in irradiated cells. RESULTS TIPRL1 expression was increased in tumor versus non-tumor tissue, with high tumoral TIPRL1 expression associating with lower locoregional control and decreased survival of RT-treated patients. TIPRL1 deletion in HNSCC cells resulted in increased RT sensitivity, a faster but prolonged cell cycle arrest, increased micronuclei formation and an altered proteome-wide DDR. Upon irradiation, ATM phosphorylates TIPRL1 at Ser265. A non-phospho Ser265Ala mutant could not rescue the increased radiosensitivity phenotype of TIPRL1-depleted cells. While binding to PP2A-like phosphatases was confirmed, DNA-dependent protein kinase (DNA-PKcs), RAD51 recombinase and nucleosomal histones were identified as novel TIPRL1 interactors. Histone binding, although stimulated by RT, was adversely affected by TIPRL1 Ser265 phosphorylation. CONCLUSIONS Our findings underscore a clinically relevant role for TIPRL1 and its ATM-dependent phosphorylation in RT resistance through modulation of the DDR, highlighting its potential as a new HNSCC predictive marker and therapeutic target.
Collapse
Affiliation(s)
- Célie Cokelaere
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
| | - Rüveyda Dok
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Emanuela E Cortesi
- Translational Cell & Tissue Research, Department of Imaging & Pathology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Peihua Zhao
- VIB Laboratory of Mechanisms of Cell Transformation, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Anna Sablina
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
- VIB Laboratory of Mechanisms of Cell Transformation, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Sandra Nuyts
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
- SybioMA, Proteomics Core Facility, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), B-3000, Leuven, Belgium.
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium.
| |
Collapse
|
13
|
Hui X, Li L, Xiong W, Liu Y, Li H, Zhang H, Zhao S, Zhang Y. High PPP4C expression predicts poor prognosis in diffuse large B-cell lymphoma. Clin Exp Med 2024; 24:89. [PMID: 38683255 PMCID: PMC11058967 DOI: 10.1007/s10238-024-01356-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
The significance of Protein phosphatase 4 catalytic subunit (PPP4C) in diffuse large B-cell lymphoma (DLBCL) prognosis is not well understood. This work aimed to investigate the expression of PPP4C in DLBCL, investigate the correlation between PPP4C expression and clinicopathological parameters, and assess the prognostic significance of PPP4C. The mRNA expression of PPP4C was investigated using data from TCGA and GEO. To further analyze PPP4C expression, immunohistochemistry was performed on tissue microarray samples. Correlation analysis between clinicopathological parameters and PPP4C expression was conducted using Pearson's chi-square test or Fisher's exact test. Univariate and multivariate Cox hazard models were utilized to determine the prognostic significance of clinicopathological features and PPP4C expression. Additionally, survival analysis was performed using Kaplan-Meier survival curves. In both TCGA and GEO datasets, we identified higher mRNA levels of PPP4C in tumor tissues compared to normal tissues. Upon analysis of various clinicopathological features of DLBCL, we observed a correlation between high PPP4C expression and ECOG score (P = 0.003). Furthermore, according to a Kaplan-Meier survival analysis, patients with DLBCL who exhibit high levels of PPP4C had worse overall survival (P = 0.001) and progression-free survival (P = 0.002). PPP4C was shown to be an independent predictive factor for OS and PFS in DLBCL by univariate and multivariate analysis (P = 0.011 and P = 0.040). This study's findings indicate that high expression of PPP4C is linked to a poor prognosis for DLBCL and may function as an independent prognostic factors.
Collapse
Affiliation(s)
- Xue Hui
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Liru Li
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Wenjing Xiong
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Yue Liu
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Hongbin Li
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Han Zhang
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Shu Zhao
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| | - Yue Zhang
- Department of Clinical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
14
|
Burigotto M, Vigorito V, Gliech C, Mattivi A, Ghetti S, Bisio A, Lolli G, Holland AJ, Fava LL. PLK1 promotes the mitotic surveillance pathway by controlling cytosolic 53BP1 availability. EMBO Rep 2023; 24:e57234. [PMID: 37888778 PMCID: PMC10702821 DOI: 10.15252/embr.202357234] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
53BP1 acts at the crossroads between DNA repair and p53-mediated stress response. With its interactors p53 and USP28, it is part of the mitotic surveillance (or mitotic stopwatch) pathway (MSP), a sensor that monitors the duration of cell division, promoting p53-dependent cell cycle arrest when a critical time threshold is surpassed. Here, we show that Polo-like kinase 1 (PLK1) activity is essential for the time-dependent release of 53BP1 from kinetochores. PLK1 inhibition, which leads to 53BP1 persistence at kinetochores, prevents cytosolic 53BP1 association with p53 and results in a blunted MSP. Strikingly, the identification of CENP-F as the kinetochore docking partner of 53BP1 enabled us to show that measurement of mitotic timing by the MSP does not take place at kinetochores, as perturbing CENP-F-53BP1 binding had no measurable impact on the MSP. Taken together, we propose that PLK1 supports the MSP by generating a cytosolic pool of 53BP1 and that an unknown cytosolic mechanism enables the measurement of mitotic duration.
Collapse
Affiliation(s)
- Matteo Burigotto
- Armenise‐Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
- Present address:
Comprehensive Cancer Centre, School of Cancer and Pharmaceutical SciencesKing's CollegeLondonUK
- Present address:
Organelle Dynamics LaboratoryThe Francis Crick InstituteLondonUK
| | - Vincenza Vigorito
- Armenise‐Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
| | - Colin Gliech
- Department of Molecular Biology and GeneticsJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Alessia Mattivi
- Armenise‐Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
| | - Sabrina Ghetti
- Armenise‐Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
- Present address:
Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Alessandra Bisio
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
| | - Graziano Lolli
- Laboratory of Protein Crystallography and Structure‐Based Drug Design, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
| | | | - Luca L Fava
- Armenise‐Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology – CIBIOUniversity of TrentoTrentoItaly
| |
Collapse
|
15
|
Park J, Lee DH. Protein phosphatase 4 dephosphorylates phosphofructokinase-1 to regulate its enzymatic activity. BMB Rep 2023; 56:618-623. [PMID: 37605615 PMCID: PMC10689085 DOI: 10.5483/bmbrep.2023-0065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/31/2023] [Accepted: 08/14/2023] [Indexed: 05/07/2025] Open
Abstract
Most cancer cells utilize glucose at a high rate to produce energyand precursors for the biosynthesis of macromolecules such as lipids, proteins, and nucleic acids. This phenomenon is called the Warburg effect or aerobic glycolysis- this distinct characteristic is an attractive target for developing anticancer drugs. Here, we found that Phosphofructokinase-1 (PFK-1) is a substrate of the Protein Phosphatase 4 catalytic subunit (PP4C)/PP4 regulatory subunit 1 (PP4R1) complex by using immunoprecipitation and in vitro assay. While manipulation of PP4C/PP4R1 does not have a critical impact on PFK-1 expression, the absence of the PP4C/PP4R1 complex increases PFK-1 activity. Although PP4C depletion or overexpression does not cause a dramatic change in the overall glycolytic rate, PP4R1 depletion induces a considerable increase in both basal and compensatory glycolytic rates, as well as the oxygen consumption rate, indicating oxidative phosphorylation. Collectively, the PP4C/PP4R1 complex regulates PFK-1 activity by reversing its phosphorylation and is a promising candidate for treating glycolytic disorders and cancers. Targeting PP4R1 could be a more efficient and safer strategy to avoid pleiotropic effects than targeting PP4C directly. [BMB Reports 2023; 56(11): 618-623].
Collapse
Affiliation(s)
- Jaehong Park
- School of Biological Sciences and Biotechnology Graduate School, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Hyun Lee
- Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, Korea
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
16
|
Park J, Lee DH. Protein phosphatase 4 dephosphorylates phosphofructokinase-1 to regulate its enzymatic activity. BMB Rep 2023; 56:618-623. [PMID: 37605615 PMCID: PMC10689085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/31/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023] Open
Abstract
Most cancer cells utilize glucose at a high rate to produce energyand precursors for the biosynthesis of macromolecules such as lipids, proteins, and nucleic acids. This phenomenon is called the Warburg effect or aerobic glycolysis- this distinct characteristic is an attractive target for developing anticancer drugs. Here, we found that Phosphofructokinase-1 (PFK-1) is a substrate of the Protein Phosphatase 4 catalytic subunit (PP4C)/PP4 regulatory subunit 1 (PP4R1) complex by using immunoprecipitation and in vitro assay. While manipulation of PP4C/PP4R1 does not have a critical impact on PFK-1 expression, the absence of the PP4C/PP4R1 complex increases PFK-1 activity. Although PP4C depletion or overexpression does not cause a dramatic change in the overall glycolytic rate, PP4R1 depletion induces a considerable increase in both basal and compensatory glycolytic rates, as well as the oxygen consumption rate, indicating oxidative phosphorylation. Collectively, the PP4C/PP4R1 complex regulates PFK-1 activity by reversing its phosphorylation and is a promising candidate for treating glycolytic disorders and cancers. Targeting PP4R1 could be a more efficient and safer strategy to avoid pleiotropic effects than targeting PP4C directly. [BMB Reports 2023; 56(11): 618-623].
Collapse
Affiliation(s)
- Jaehong Park
- School of Biological Sciences and Biotechnology Graduate School, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Hyun Lee
- Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, Korea
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
17
|
Réthi-Nagy Z, Ábrahám E, Sinka R, Juhász S, Lipinszki Z. Protein Phosphatase 4 Is Required for Centrobin Function in DNA Damage Repair. Cells 2023; 12:2219. [PMID: 37759442 PMCID: PMC10526779 DOI: 10.3390/cells12182219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Genome stability in human cells relies on the efficient repair of double-stranded DNA breaks, which is mainly achieved by homologous recombination (HR). Among the regulators of various cellular functions, Protein phosphatase 4 (PP4) plays a pivotal role in coordinating cellular response to DNA damage. Meanwhile, Centrobin (CNTRB), initially recognized for its association with centrosomal function and microtubule dynamics, has sparked interest due to its potential contribution to DNA repair processes. In this study, we investigate the involvement of PP4 and its interaction with CNTRB in HR-mediated DNA repair in human cells. Employing a range of experimental strategies, we investigate the physical interaction between PP4 and CNTRB and shed light on the importance of two specific motifs in CNTRB, the PP4-binding FRVP and the ATR kinase recognition SQ sequences, in the DNA repair process. Moreover, we examine cells depleted of PP4 or CNTRB and cells harboring FRVP and SQ mutations in CNTRB, which result in similar abnormal chromosome morphologies. This phenomenon likely results from the impaired resolution of Holliday junctions, which serve as crucial intermediates in HR. Taken together, our results provide new insights into the intricate mechanisms of PP4 and CNTRB-regulated HR repair and their interrelation.
Collapse
Affiliation(s)
- Zsuzsánna Réthi-Nagy
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, H-6726 Szeged, Hungary
| | - Edit Ábrahám
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| | - Rita Sinka
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary;
| | - Szilvia Juhász
- Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| | - Zoltán Lipinszki
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| |
Collapse
|
18
|
Gelot C, Kovacs MT, Miron S, Mylne E, Haan A, Boeffard-Dosierre L, Ghouil R, Popova T, Dingli F, Loew D, Guirouilh-Barbat J, Del Nery E, Zinn-Justin S, Ceccaldi R. Polθ is phosphorylated by PLK1 to repair double-strand breaks in mitosis. Nature 2023; 621:415-422. [PMID: 37674080 PMCID: PMC10499603 DOI: 10.1038/s41586-023-06506-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 08/01/2023] [Indexed: 09/08/2023]
Abstract
DNA double-strand breaks (DSBs) are deleterious lesions that challenge genome integrity. To mitigate this threat, human cells rely on the activity of multiple DNA repair machineries that are tightly regulated throughout the cell cycle1. In interphase, DSBs are mainly repaired by non-homologous end joining and homologous recombination2. However, these pathways are completely inhibited in mitosis3-5, leaving the fate of mitotic DSBs unknown. Here we show that DNA polymerase theta6 (Polθ) repairs mitotic DSBs and thereby maintains genome integrity. In contrast to other DSB repair factors, Polθ function is activated in mitosis upon phosphorylation by Polo-like kinase 1 (PLK1). Phosphorylated Polθ is recruited by a direct interaction with the BRCA1 C-terminal domains of TOPBP1 to mitotic DSBs, where it mediates joining of broken DNA ends. Loss of Polθ leads to defective repair of mitotic DSBs, resulting in a loss of genome integrity. This is further exacerbated in cells that are deficient in homologous recombination, where loss of mitotic DSB repair by Polθ results in cell death. Our results identify mitotic DSB repair as the underlying cause of synthetic lethality between Polθ and homologous recombination. Together, our findings reveal the critical importance of mitotic DSB repair in the maintenance of genome integrity.
Collapse
Affiliation(s)
- Camille Gelot
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | | | - Simona Miron
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
| | - Emilie Mylne
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Alexis Haan
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Liza Boeffard-Dosierre
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
| | - Rania Ghouil
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
| | - Tatiana Popova
- INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, PSL Research University, Institut Curie, Paris, France
| | - Florent Dingli
- CurieCoreTech Mass Spectrometry Proteomics, Institut Curie, PSL Research University, Paris, France
| | - Damarys Loew
- CurieCoreTech Mass Spectrometry Proteomics, Institut Curie, PSL Research University, Paris, France
| | - Josée Guirouilh-Barbat
- Université de Paris, INSERM U1016, UMR 8104 CNRS, Equipe Labellisée Ligue Nationale Contre le Cancer, Institut Cochin, Paris, France
| | - Elaine Del Nery
- Department of Translational Research-Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), PSL Research University, Institut Curie, Paris, France
| | - Sophie Zinn-Justin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
| | - Raphael Ceccaldi
- INSERM U830, PSL Research University, Institut Curie, Paris, France.
| |
Collapse
|
19
|
Brambati A, Sacco O, Porcella S, Heyza J, Kareh M, Schmidt JC, Sfeir A. RHINO directs MMEJ to repair DNA breaks in mitosis. Science 2023; 381:653-660. [PMID: 37440612 PMCID: PMC10561558 DOI: 10.1126/science.adh3694] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
Nonhomologous end-joining (NHEJ) and homologous recombination (HR) are the primary pathways for repairing DNA double-strand breaks (DSBs) during interphase, whereas microhomology-mediated end-joining (MMEJ) has been regarded as a backup mechanism. Through CRISPR-Cas9-based synthetic lethal screens in cancer cells, we identified subunits of the 9-1-1 complex (RAD9A-RAD1-HUS1) and its interacting partner, RHINO, as crucial MMEJ factors. We uncovered an unexpected function for RHINO in restricting MMEJ to mitosis. RHINO accumulates in M phase, undergoes Polo-like kinase 1 (PLK1) phosphorylation, and interacts with polymerase θ (Polθ), enabling its recruitment to DSBs for subsequent repair. Additionally, we provide evidence that MMEJ activity in mitosis repairs persistent DSBs that originate in S phase. Our findings offer insights into the synthetic lethal relationship between the genes POLQ and BRCA1 and BRAC2 and the synergistic effect of Polθ and poly(ADP-ribose) polymerase (PARP) inhibitors.
Collapse
Affiliation(s)
- Alessandra Brambati
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Olivia Sacco
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Sarina Porcella
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Joshua Heyza
- Institute for Quantitative Health Sciences and Engineering, Michigan State University; East Lansing, MI, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University; East Lansing, MI, USA
| | - Mike Kareh
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Jens C. Schmidt
- Institute for Quantitative Health Sciences and Engineering, Michigan State University; East Lansing, MI, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University; East Lansing, MI, USA
| | - Agnel Sfeir
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| |
Collapse
|
20
|
Raja R, Wu C, Bassoy EY, Rubino TE, Utagawa EC, Magtibay PM, Butler KA, Curtis M. PP4 inhibition sensitizes ovarian cancer to NK cell-mediated cytotoxicity via STAT1 activation and inflammatory signaling. J Immunother Cancer 2022; 10:jitc-2022-005026. [PMID: 36564125 PMCID: PMC9791393 DOI: 10.1136/jitc-2022-005026] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Increased infiltration of T cells into ovarian tumors has been repeatedly shown to be predictive of enhanced patient survival. However, despite the evidence of an active immune response in ovarian cancer (OC), the frequency of responses to immune checkpoint blockade (ICB) therapy in OC is much lower than other cancer types. Recent studies have highlighted that deficiencies in the DNA damage response (DDR) can drive increased genomic instability and tumor immunogenicity, which leads to enhanced responses to ICB. Protein phosphatase 4 (PP4) is a critical regulator of the DDR; however, its potential role in antitumor immunity is currently unknown. RESULTS Our results show that the PP4 inhibitor, fostriecin, combined with carboplatin leads to increased carboplatin sensitivity, DNA damage, and micronuclei formation. Using multiple OC cell lines, we show that PP4 inhibition or PPP4C knockdown combined with carboplatin triggers inflammatory signaling via Nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 1 (STAT1) activation. This resulted in increased expression of the pro-inflammatory cytokines and chemokines: CCL5, CXCL10, and IL-6. In addition, IFNB1 expression was increased suggesting activation of the type I interferon response. Conditioned media from OC cells treated with the combination of PP4 inhibitor and carboplatin significantly increased migration of both CD8 T cell and natural killer (NK) cells over carboplatin treatment alone. Knockdown of stimulator of interferon genes (STING) in OC cells significantly abrogated the increase in CD8 T-cell migration induced by PP4 inhibition. Co-culture of NK-92 cells and OC cells with PPP4C or PPP4R3B knockdown resulted in strong induction of NK cell interferon-γ, increased degranulation, and increased NK cell-mediated cytotoxicity against OC cells. Stable knockdown of PP4C in a syngeneic, immunocompetent mouse model of OC resulted in significantly reduced tumor growth in vivo. Tumors with PP4C knockdown had increased infiltration of NK cells, NK T cells, and CD4+ T cells. Addition of low dose carboplatin treatment led to increased CD8+ T-cell infiltration in PP4C knockdown tumors as compared with the untreated PP4C knockdown tumors. CONCLUSIONS Our work has identified a role for PP4 inhibition in promoting inflammatory signaling and enhanced immune cell effector function. These findings support the further investigation of PP4 inhibitors to enhance chemo-immunotherapy for OC treatment.
Collapse
Affiliation(s)
- Remya Raja
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Christopher Wu
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Esen Yonca Bassoy
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Thomas E Rubino
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Emma C Utagawa
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA
| | - Paul M Magtibay
- Department of Gynecology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Kristina A Butler
- Department of Gynecology, Mayo Clinic, Scottsdale, Arizona, USA,College of Medicine and Science, Mayo Clinic, Scottsdale, Arizona, USA
| | - Marion Curtis
- Department of Immunology, Mayo Clinic Scottsdale, Scottsdale, Arizona, USA,College of Medicine and Science, Mayo Clinic, Scottsdale, Arizona, USA,Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
21
|
Rass E, Willaume S, Bertrand P. 53BP1: Keeping It under Control, Even at a Distance from DNA Damage. Genes (Basel) 2022; 13:genes13122390. [PMID: 36553657 PMCID: PMC9778356 DOI: 10.3390/genes13122390] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Double-strand breaks (DSBs) are toxic lesions that can be generated by exposure to genotoxic agents or during physiological processes, such as during V(D)J recombination. The repair of these DSBs is crucial to prevent genomic instability and to maintain cellular homeostasis. Two main pathways participate in repairing DSBs, namely, non-homologous end joining (NHEJ) and homologous recombination (HR). The P53-binding protein 1 (53BP1) plays a pivotal role in the choice of DSB repair mechanism, promotes checkpoint activation and preserves genome stability upon DSBs. By preventing DSB end resection, 53BP1 promotes NHEJ over HR. Nonetheless, the balance between DSB repair pathways remains crucial, as unscheduled NHEJ or HR events at different phases of the cell cycle may lead to genomic instability. Therefore, the recruitment of 53BP1 to chromatin is tightly regulated and has been widely studied. However, less is known about the mechanism regulating 53BP1 recruitment at a distance from the DNA damage. The present review focuses on the mechanism of 53BP1 recruitment to damage and on recent studies describing novel mechanisms keeping 53BP1 at a distance from DSBs.
Collapse
Affiliation(s)
- Emilie Rass
- Université Paris Cité, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Université Paris-Saclay, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Correspondence:
| | - Simon Willaume
- Université Paris Cité, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Université Paris-Saclay, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
| | - Pascale Bertrand
- Université Paris Cité, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Université Paris-Saclay, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
| |
Collapse
|
22
|
Temporal phosphoproteomics reveals WEE1-dependent control of 53BP1 pathway. iScience 2022; 26:105806. [PMID: 36632060 PMCID: PMC9827073 DOI: 10.1016/j.isci.2022.105806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/29/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Wee1-like protein kinase (WEE1) restrains activities of cyclin-dependent kinases (CDKs) in S and G2 phase. Inhibition of WEE1 evokes drastic increase in CDK activity, which perturbs replication dynamics and compromises cell cycle checkpoints. Notably, WEE1 inhibitors such as adavosertib are tested in cancer treatment trials; however, WEE1-regulated phosphoproteomes and their dynamics have not been systematically investigated. In this study, we identified acute time-resolved alterations in the cellular phosphoproteome following WEE1 inhibition with adavosertib. These treatments acutely elevated CDK activities with distinct phosphorylation dynamics revealing more than 600 potential uncharacterized CDK sites. Moreover, we identified a major role for WEE1 in controlling CDK-dependent phosphorylation of multiple clustered sites in the key DNA repair factors MDC1, 53BP1, and RIF1. Functional analysis revealed that WEE1 fine-tunes CDK activities to permit recruitment of 53BP1 to chromatin. Thus, our findings uncover WEE1-controlled targets and pathways with translational potential for the clinical application of WEE1 inhibitors.
Collapse
|
23
|
Lei T, Du S, Peng Z, Chen L. Multifaceted regulation and functions of 53BP1 in NHEJ‑mediated DSB repair (Review). Int J Mol Med 2022; 50:90. [PMID: 35583003 PMCID: PMC9162042 DOI: 10.3892/ijmm.2022.5145] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/29/2022] [Indexed: 12/02/2022] Open
Abstract
The repair of DNA double-strand breaks (DSBs) is crucial for the preservation of genomic integrity and the maintenance of cellular homeostasis. Non-homologous DNA end joining (NHEJ) is the predominant repair mechanism for any type of DNA DSB during the majority of the cell cycle. NHEJ defects regulate tumor sensitivity to ionizing radiation and anti-neoplastic agents, resulting in immunodeficiencies and developmental abnormalities in malignant cells. p53-binding protein 1 (53BP1) is a key mediator involved in DSB repair, which functions to maintain a balance in the repair pathway choices and in preserving genomic stability. 53BP1 promotes DSB repair via NHEJ and antagonizes DNA end overhang resection. At present, novel lines of evidence have revealed the molecular mechanisms underlying the recruitment of 53BP1 and DNA break-responsive effectors to DSB sites, and the promotion of NHEJ-mediated DSB repair via 53BP1, while preventing homologous recombination. In the present review article, recent advances made in the elucidation of the structural and functional characteristics of 53BP1, the mechanisms of 53BP1 recruitment and interaction with the reshaping of the chromatin architecture around DSB sites, the post-transcriptional modifications of 53BP1, and the up- and downstream pathways of 53BP1 are discussed. The present review article also focuses on the application perspectives, current challenges and future directions of 53BP1 research.
Collapse
Affiliation(s)
- Tiantian Lei
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R. China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Zhe Peng
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R. China
| | - Lin Chen
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, P.R. China
| |
Collapse
|
24
|
Dong MZ, Ouyang YC, Gao SC, Ma XS, Hou Y, Schatten H, Wang ZB, Sun QY. PPP4C facilitates homologous recombination DNA repair by dephosphorylating PLK1 during early embryo development. Development 2022; 149:dev200351. [PMID: 35546066 DOI: 10.1242/dev.200351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/24/2022] [Indexed: 12/17/2023]
Abstract
Mammalian early embryo cells have complex DNA repair mechanisms to maintain genomic integrity, and homologous recombination (HR) plays the main role in response to double-strand DNA breaks (DSBs) in these cells. Polo-like kinase 1 (PLK1) participates in the HR process and its overexpression has been shown to occur in a variety of human cancers. Nevertheless, the regulatory mechanism of PLK1 remains poorly understood, especially during the S and G2 phase. Here, we show that protein phosphatase 4 catalytic subunit (PPP4C) deletion causes severe female subfertility due to accumulation of DNA damage in oocytes and early embryos. PPP4C dephosphorylated PLK1 at the S137 site, negatively regulating its activity in the DSB response in early embryonic cells. Depletion of PPP4C induced sustained activity of PLK1 when cells exhibited DNA lesions that inhibited CHK2 and upregulated the activation of CDK1, resulting in inefficient loading of the essential HR factor RAD51. On the other hand, when inhibiting PLK1 in the S phase, DNA end resection was restricted. These results demonstrate that PPP4C orchestrates the switch between high-PLK1 and low-PLK1 periods, which couple the checkpoint to HR.
Collapse
Affiliation(s)
- Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shi-Cai Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xue-Shan Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| |
Collapse
|
25
|
Salem Wehbe L, Barakat D, Acker A, El Khoury R, Reichhart JM, Matt N, El Chamy L. Protein Phosphatase 4 Negatively Regulates the Immune Deficiency-NF-κB Pathway during the Drosophila Immune Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1616-1626. [PMID: 34452932 PMCID: PMC7616922 DOI: 10.4049/jimmunol.1901497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 07/07/2021] [Indexed: 12/31/2022]
Abstract
The evolutionarily conserved immune deficiency (IMD) signaling pathway shields Drosophila against bacterial infections. It regulates the expression of antimicrobial peptides encoding genes through the activation of the NF-κB transcription factor Relish. Tight regulation of the signaling cascade ensures a balanced immune response, which is otherwise highly harmful. Several phosphorylation events mediate intracellular progression of the IMD pathway. However, signal termination by dephosphorylation remains largely elusive. Here, we identify the highly conserved protein phosphatase 4 (PP4) complex as a bona fide negative regulator of the IMD pathway. RNA interference-mediated gene silencing of PP4-19c, PP4R2, and Falafel, which encode the catalytic and regulatory subunits of the phosphatase complex, respectively, caused a marked upregulation of bacterial-induced antimicrobial peptide gene expression in both Drosophila melanogaster S2 cells and adult flies. Deregulated IMD signaling is associated with reduced lifespan of PP4-deficient flies in the absence of any infection. In contrast, flies overexpressing this phosphatase are highly sensitive to bacterial infections. Altogether, our results highlight an evolutionarily conserved function of PP4c in the regulation of NF-κB signaling from Drosophila to mammals.
Collapse
Affiliation(s)
- Layale Salem Wehbe
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | - Dana Barakat
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | - Adrian Acker
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
| | - Rita El Khoury
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | | | - Nicolas Matt
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
| | - Laure El Chamy
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| |
Collapse
|
26
|
De Marco Zompit M, Stucki M. Mechanisms of genome stability maintenance during cell division. DNA Repair (Amst) 2021; 108:103215. [PMID: 34455186 DOI: 10.1016/j.dnarep.2021.103215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 01/12/2023]
Abstract
During mitosis, chromosomes undergo extensive structural changes resulting in the formation of compact cylindrical bodies and in the termination of the bulk of DNA-dependent metabolic activities. Therefore, DNA lesions that interfere with processes such as DNA replication and transcription in interphase are not expected to pose a major threat to genome stability in mitosis. There are, however, a few exceptions. DNA replication and repair intermediates that physically interconnect the sister chromatids jeopardize faithful chromosome segregation and need to be resolved before the onset of anaphase. In addition, dicentric chromosomes can form chromatin bridges and induce breakage-fusion-breakage cycles with dire consequences for genome stability. Finally, chromosome breaks that escape the G2/M DNA damage checkpoint or emerge early in mitosis may result in lagging acentric DNA fragments that mis-segregate and form micronuclei when cells exit from mitosis. Both chromatin bridges and micronuclei are potential sources of a mutational cascade that results in massive chromosomal instability and significantly contributes to genomic complexity. Here, we review recent progress in our understanding of the origins and consequences of chromosome bridges and micronuclei and the mechanisms by which cells suppress them.
Collapse
Affiliation(s)
- Mara De Marco Zompit
- Department of Gynecology, University Hospital Zurich, University of Zurich, Wagistrasse 14, CH-8952 Schlieren, Switzerland
| | - Manuel Stucki
- Department of Gynecology, University Hospital Zurich, University of Zurich, Wagistrasse 14, CH-8952 Schlieren, Switzerland.
| |
Collapse
|
27
|
Etourneaud L, Moussa A, Rass E, Genet D, Willaume S, Chabance-Okumura C, Wanschoor P, Picotto J, Thézé B, Dépagne J, Veaute X, Dizet E, Busso D, Barascu A, Irbah L, Kortulewski T, Campalans A, Le Chalony C, Zinn-Justin S, Scully R, Pennarun G, Bertrand P. Lamin B1 sequesters 53BP1 to control its recruitment to DNA damage. SCIENCE ADVANCES 2021; 7:eabb3799. [PMID: 34452908 PMCID: PMC8397269 DOI: 10.1126/sciadv.abb3799] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/07/2021] [Indexed: 05/09/2023]
Abstract
Double-strand breaks (DSBs) are harmful lesions and a major cause of genome instability. Studies have suggested a link between the nuclear envelope and the DNA damage response. Here, we show that lamin B1, a major component of the nuclear envelope, interacts directly with 53BP1 protein, which plays a pivotal role in the DSB repair. This interaction is dissociated after DNA damage. Lamin B1 overexpression impedes 53BP1 recruitment to DNA damage sites and leads to a persistence of DNA damage, a defect in nonhomologous end joining and an increased sensitivity to DSBs. The identification of interactions domains between lamin B1 and 53BP1 allows us to demonstrate that the defect of 53BP1 recruitment and the DSB persistence upon lamin B1 overexpression are due to sequestration of 53BP1 by lamin B1. This study highlights lamin B1 as a factor controlling the recruitment of 53BP1 to DNA damage sites upon injury.
Collapse
Affiliation(s)
- Laure Etourneaud
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Angela Moussa
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Emilie Rass
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Diane Genet
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Simon Willaume
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Caroline Chabance-Okumura
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Paul Wanschoor
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Julien Picotto
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Benoît Thézé
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Jordane Dépagne
- Genetic Engineering and Expression Platform (CIGEX), iRCM, DRF, CEA, Fontenay-aux-Roses, France
| | - Xavier Veaute
- Genetic Engineering and Expression Platform (CIGEX), iRCM, DRF, CEA, Fontenay-aux-Roses, France
| | - Eléa Dizet
- Genetic Engineering and Expression Platform (CIGEX), iRCM, DRF, CEA, Fontenay-aux-Roses, France
| | - Didier Busso
- Genetic Engineering and Expression Platform (CIGEX), iRCM, DRF, CEA, Fontenay-aux-Roses, France
| | - Aurélia Barascu
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Lamya Irbah
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- Imaging platform, iRCM, DRF, CEA, F-92265 Fontenay-aux-Roses, France
| | - Thierry Kortulewski
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "Radiopathology" Team, iRCM/IBFJ, DRF, CEA, France
| | - Anna Campalans
- Université de Paris and Université Paris Saclay, iRCM/IBFJ, CEA, UMR Stabilité Génétique Cellules Souches et Radiations, "Genetic Instability Research" Team, F-92265 Fontenay-aux-Roses, France
| | - Catherine Le Chalony
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Sophie Zinn-Justin
- Laboratory of Structural Biology and Radiobiology, Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, F-91190 Gif-sur-Yvette, France
| | - Ralph Scully
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Gaëlle Pennarun
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| | - Pascale Bertrand
- Université de Paris and Université Paris Saclay, INSERM, iRCM/IBFJ, CEA, UMR Stabilité Génétique, Cellules Souches et Radiations, F-92265 Fontenay-aux-Roses, France.
- "DNA Repair and Ageing" Team, iRCM/IBFJ, DRF, CEA, France
| |
Collapse
|
28
|
p85β alters response to EGFR inhibitor in ovarian cancer through p38 MAPK-mediated regulation of DNA repair. Neoplasia 2021; 23:718-730. [PMID: 34144267 PMCID: PMC8220107 DOI: 10.1016/j.neo.2021.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
Abstract
EGFR signaling promotes ovarian cancer tumorigenesis, and high EGFR expression correlates with poor prognosis. However, EGFR inhibitors alone have demonstrated limited clinical benefit for ovarian cancer patients, owing partly to tumor resistance and the lack of predictive biomarkers. Cotargeting EGFR and the PI3K pathway has been previously shown to yield synergistic antitumor effects in ovarian cancer. Therefore, we reasoned that PI3K may affect cellular response to EGFR inhibition. In this study, we revealed PI3K isoform-specific effects on the sensitivity of ovarian cancer cells to the EGFR inhibitor erlotinib. Gene silencing of PIK3CA (p110α) and PIK3CB (p110β) rendered cells more susceptible to erlotinib. In contrast, low expression of PIK3R2 (p85β) was associated with erlotinib resistance. Depletion of PIK3R2, but not PIK3CA or PIK3CB, led to increased DNA damage and reduced level of the nonhomologous end joining DNA repair protein BRD4. Intriguingly, these defects in DNA repair were reversed upon erlotinib treatment, which caused activation and nuclear import of p38 MAPK to promote DNA repair with increased protein levels of 53BP1 and BRD4 and foci formation of 53BP1. Remarkably, inhibition of p38 MAPK or BRD4 re-sensitized PIK3R2-depleted cells to erlotinib. Collectively, these data suggest that p38 MAPK activation and the subsequent DNA repair serve as a resistance mechanism to EGFR inhibitor. Combined inhibition of EGFR and p38 MAPK or DNA repair may maximize the therapeutic potential of EGFR inhibitor in ovarian cancer.
Collapse
|
29
|
Willaume S, Rass E, Fontanilla-Ramirez P, Moussa A, Wanschoor P, Bertrand P. A Link between Replicative Stress, Lamin Proteins, and Inflammation. Genes (Basel) 2021; 12:genes12040552. [PMID: 33918867 PMCID: PMC8070205 DOI: 10.3390/genes12040552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/23/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Double-stranded breaks (DSB), the most toxic DNA lesions, are either a consequence of cellular metabolism, programmed as in during V(D)J recombination, or induced by anti-tumoral therapies or accidental genotoxic exposure. One origin of DSB sources is replicative stress, a major source of genome instability, especially when the integrity of the replication forks is not properly guaranteed. To complete stalled replication, restarting the fork requires complex molecular mechanisms, such as protection, remodeling, and processing. Recently, a link has been made between DNA damage accumulation and inflammation. Indeed, defects in DNA repair or in replication can lead to the release of DNA fragments in the cytosol. The recognition of this self-DNA by DNA sensors leads to the production of inflammatory factors. This beneficial response activating an innate immune response and destruction of cells bearing DNA damage may be considered as a novel part of DNA damage response. However, upon accumulation of DNA damage, a chronic inflammatory cellular microenvironment may lead to inflammatory pathologies, aging, and progression of tumor cells. Progress in understanding the molecular mechanisms of DNA damage repair, replication stress, and cytosolic DNA production would allow to propose new therapeutical strategies against cancer or inflammatory diseases associated with aging. In this review, we describe the mechanisms involved in DSB repair, the replicative stress management, and its consequences. We also focus on new emerging links between key components of the nuclear envelope, the lamins, and DNA repair, management of replicative stress, and inflammation.
Collapse
|
30
|
Wang G, Chen X, Wang N, Xiao Y, Shu S, Alsayed AMM, Liu L, Ma Y, Liu P, Zhang Q, Chen X, Liu Z, Zheng X. The discovery of novel sanjuanolide derivatives as chemotherapeutic agents targeting castration-resistant prostate cancer. Bioorg Chem 2021; 111:104880. [PMID: 33839585 DOI: 10.1016/j.bioorg.2021.104880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/20/2022]
Abstract
There remains a critical need for more effective therapies for the treatment of castration-resistant prostate cancer (CRPC), which is the leading cause of death in patients with prostate cancer. In this study, a series of sanjuanolide derivatives were designed, synthesized and evaluated as potential anti-CRPC agents. Most of the compounds had excellent selectivity for CRPC cells with IC50 values < 20 µM. Moreover, minimal side effects on human normal hepatic MIHA cells and normal prostatic stromal myofibroblast WPMY-1 cells were observed, with IC50 > 100 µM. The representative compound S07 slowed down the proliferative rate of CRPC cells, promoted cell apoptosis and caused G2/M phase accumulation, as well as G1/G0 phase reduction. Further mechanistic studies showed that S07 treatment triggered intense DNA damage and provoked strong DNA damage response in a dose-dependent manner. These findings suggested that sanjuanolide derivatives, especially S07, selectively induced CRPC cell death by triggering intense DNA damage and DNA damage response.
Collapse
Affiliation(s)
- Guangbao Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Xiaojing Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Nan Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yunbei Xiao
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Sheng Shu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Ali Mohammed Mohammed Alsayed
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Lu Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yue Ma
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Peng Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Qianwen Zhang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Xiangjuan Chen
- Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518000, China.
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
31
|
Jiang Y, Dong Y, Luo Y, Jiang S, Meng FL, Tan M, Li J, Zang Y. AMPK-mediated phosphorylation on 53BP1 promotes c-NHEJ. Cell Rep 2021; 34:108713. [PMID: 33596428 DOI: 10.1016/j.celrep.2021.108713] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/12/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is an energy sensor that plays roles in multiple biological processes beyond metabolism. Several studies have suggested that AMPK is involved in the DNA damage response (DDR), but the mechanisms remain unclear. Herein, we demonstrate that AMPK promotes classic non-homologous end joining (c-NHEJ) in double-strand break (DSB) repair through recruiting a key chromatin-based mediator named p53-binding protein 1 (53BP1), which facilitates the end joining of distal DNA ends during DDR. We find that the interaction of AMPK and 53BP1 spatially occurs under DSB stress. In the context of DSBs, AMPK directly phosphorylates 53BP1 at Ser1317 and promotes 53BP1 recruitment during DDR for an efficient c-NHEJ, thus maintaining genomic stability and diversity of the immune repertoire. Taken together, our study demonstrates that AMPK is a regulator of 53BP1 and controls c-NHEJ choice by phospho-regulation.
Collapse
Affiliation(s)
- Yuejing Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Yifeng Luo
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China; State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Shangwen Jiang
- Chemical Proteomics Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fei-Long Meng
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China; State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Minjia Tan
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China; Chemical Proteomics Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China; Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), 1 Wenhai Road, Aoshanwei, Jimo, Qingdao 266237, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| |
Collapse
|
32
|
Chen X, Wang G, Mohammed Alsayed AM, Du Z, Lu Liu, Ma Y, Liu P, Zhang Q, Chen X, Chen W, Ye F, Zheng X, Liu Z. Synthesis and biological evaluation of novel N-substituted benzamides as anti-migration agents for treatment of osteosarcoma. Eur J Med Chem 2021; 214:113203. [PMID: 33530028 DOI: 10.1016/j.ejmech.2021.113203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/25/2020] [Accepted: 01/11/2021] [Indexed: 12/20/2022]
Abstract
A novel series of novel N-substituted (indole or indazole) benzamides were synthesized, and their anti-tumor properties were evaluated. The majority of tested compounds possessed moderate cytotoxicity, but inspiringly, we verified that active compound 5d presents an astonishing advantage by inhibiting the adhesion, migration, and invasion of osteosarcoma (OS) cells in vitro. Mechanistically, we confirmed 5d inhibited the migration ability of OS cells via the expression of genes related to adhesion, migration, and invasion. This effects of 5d suggest that it can be used as a potential chemotherapeutic drug to some aggressive and/or metastatic cancers, as well as in combination with other clinical anti-cancer drugs. In turn, this could enhance the therapeutic effect or reduce the risk of cell migration.
Collapse
Affiliation(s)
- Xiaojing Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Guangbao Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Ali Mohammed Mohammed Alsayed
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Zongxuan Du
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Lu Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Yue Ma
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Peng Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Qianwen Zhang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Xianxin Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Wenbin Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China
| | - Faqing Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China.
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China.
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
33
|
Park J, Lee DH. Functional roles of protein phosphatase 4 in multiple aspects of cellular physiology: a friend and a foe. BMB Rep 2021. [PMID: 32192570 PMCID: PMC7196183 DOI: 10.5483/bmbrep.2020.53.4.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protein phosphatase 4 (PP4), one of serine/threonine phosphatases, is involved in many critical cellular pathways, including DNA damage response (DNA repair, cell cycle regulation, and apoptosis), tumorigenesis, cell migration, immune response, stem cell development, glucose metabolism, and diabetes. PP4 has been steadily studied over the past decade about wide spectrum of physiological activities in cells. Given the many vital functions in cells, PP4 has great potential to develop into the finding of key working mechanisms and effective treatments for related diseases such as cancer and diabetes. In this review, we provide an overview of the cellular and molecular mechanisms by which PP4 impacts and also discuss the functional significance of it in cell health.
Collapse
Affiliation(s)
- Jaehong Park
- School of Biological Sciences and Biotechnology Graduate School, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Hyun Lee
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju 61186; Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
34
|
Sakamoto Y, Kokuta T, Teshigahara A, Iijima K, Kitao H, Takata M, Tauchi H. Mitotic cells can repair DNA double-strand breaks via a homology-directed pathway. JOURNAL OF RADIATION RESEARCH 2021; 62:25-33. [PMID: 33009557 PMCID: PMC7779344 DOI: 10.1093/jrr/rraa095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/21/2020] [Indexed: 05/04/2023]
Abstract
The choice of repair pathways of DNA double-strand breaks (DSBs) is dependent upon the cell cycle phases. While homologous recombination repair (HRR) is active between the S and G2 phases, its involvement in mitotic DSB repair has not been examined in detail. In the present study, we developed a new reporter assay system to detect homology-directed repair (HDR), a major pathway used for HRR, in combination with an inducible DSB-generation system. As expected, the maximal HDR activity was observed in the late S phase, along with minimal activity in the G1 phase and at the G1/S boundary. Surprisingly, significant HDR activity was observed in M phase, and the repair efficiency was similar to that observed in late S phase. HDR was also confirmed in metaphase cells collected with continuous colcemid exposure. ChIP assays revealed the recruitment of RAD51 to the vicinity of DSBs in M phase. In addition, the ChIP assay for gamma-H2AX and phosphorylated DNA-PKcs indicated that a part of M-phase cells with DSBs could proceed into the next G1 phase. These results provide evidence showing that a portion of mitotic cell DSBs are undoubtedly repaired through action of the HDR repair pathway.
Collapse
Affiliation(s)
- Yuki Sakamoto
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Tetsuya Kokuta
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Ai Teshigahara
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Kenta Iijima
- Department of Biological Sciences, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
- Department of Cancer Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroyuki Kitao
- Department of Molecular Cancer Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Minoru Takata
- Radiation Biology Center, Kyoto University, Yoshida-Konoe Cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Tauchi
- Corresponding author. Department of Biological Sciences, Faculty of Science, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan. Tel: +81-29-228-8383; Fax: +81-29-228-8403;
| |
Collapse
|
35
|
Karman Z, Rethi-Nagy Z, Abraham E, Fabri-Ordogh L, Csonka A, Vilmos P, Debski J, Dadlez M, Glover DM, Lipinszki Z. Novel perspectives of target-binding by the evolutionarily conserved PP4 phosphatase. Open Biol 2020; 10:200343. [PMID: 33352067 PMCID: PMC7776573 DOI: 10.1098/rsob.200343] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Protein phosphatase 4 (PP4) is an evolutionarily conserved and essential Ser/Thr phosphatase that regulates cell division, development and DNA repair in eukaryotes. The major form of PP4, present from yeast to human, is the PP4c-R2-R3 heterotrimeric complex. The R3 subunit is responsible for substrate-recognition via its EVH1 domain. In typical EVH1 domains, conserved phenylalanine, tyrosine and tryptophan residues form the specific recognition site for their target's proline-rich sequences. Here, we identify novel binding partners of the EVH1 domain of the Drosophila R3 subunit, Falafel, and demonstrate that instead of binding to proline-rich sequences this EVH1 variant specifically recognizes atypical ligands, namely the FxxP and MxPP short linear consensus motifs. This interaction is dependent on an exclusively conserved leucine that replaces the phenylalanine invariant of all canonical EVH1 domains. We propose that the EVH1 domain of PP4 represents a new class of the EVH1 family that can accommodate low proline content sequences, such as the FxxP motif. Finally, our data implicate the conserved Smk-1 domain of Falafel in target-binding. These findings greatly enhance our understanding of the substrate-recognition mechanisms and function of PP4.
Collapse
Affiliation(s)
- Zoltan Karman
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, H‐6725, Hungary
| | - Zsuzsanna Rethi-Nagy
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, H‐6725, Hungary
| | - Edit Abraham
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
| | - Lilla Fabri-Ordogh
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
| | - Akos Csonka
- Department of Traumatology, University of Szeged, Szeged, H‐6725, Hungary
| | - Peter Vilmos
- Biological Research Centre, Institute of Genetics, Szeged, H‐6726, Hungary
| | - Janusz Debski
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Michal Dadlez
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - David M. Glover
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- California Institute of Technology, Pasadena, CA 91125, USA
| | - Zoltan Lipinszki
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
| |
Collapse
|
36
|
MacDonald KM, Benguerfi S, Harding SM. Alerting the immune system to DNA damage: micronuclei as mediators. Essays Biochem 2020; 64:753-764. [PMID: 32844183 PMCID: PMC7588664 DOI: 10.1042/ebc20200016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/01/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022]
Abstract
Healthy cells experience thousands of DNA lesions per day during normal cellular metabolism, and ionizing radiation and chemotherapeutic drugs rely on DNA damage to kill cancer cells. In response to such lesions, the DNA damage response (DDR) activates cell-cycle checkpoints, initiates DNA repair mechanisms, or promotes the clearance of irreparable cells. Work over the past decade has revealed broader influences of the DDR, involving inflammatory gene expression following unresolved DNA damage, and immune surveillance of damaged or mutated cells. Subcellular structures called micronuclei, containing broken fragments of DNA or whole chromosomes that have been isolated away from the rest of the genome, are now recognized as one mediator of DDR-associated immune recognition. Micronuclei can initiate pro-inflammatory signaling cascades, or massively degrade to invoke distinct forms of genomic instability. In this mini-review, we aim to provide an overview of the current evidence linking the DDR to activation of the immune response through micronuclei formation, identifying key areas of interest, open questions, and emerging implications.
Collapse
Affiliation(s)
- Kate M MacDonald
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Soraya Benguerfi
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Shane M Harding
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Radiation Oncology and Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Walser F, Mulder MPC, Bragantini B, Burger S, Gubser T, Gatti M, Botuyan MV, Villa A, Altmeyer M, Neri D, Ovaa H, Mer G, Penengo L. Ubiquitin Phosphorylation at Thr12 Modulates the DNA Damage Response. Mol Cell 2020; 80:423-436.e9. [PMID: 33022275 DOI: 10.1016/j.molcel.2020.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/14/2020] [Accepted: 09/12/2020] [Indexed: 11/26/2022]
Abstract
The ubiquitin system regulates the DNA damage response (DDR) by modifying histone H2A at Lys15 (H2AK15ub) and triggering downstream signaling events. Here, we find that phosphorylation of ubiquitin at Thr12 (pUbT12) controls the DDR by inhibiting the function of 53BP1, a key factor for DNA double-strand break repair by non-homologous end joining (NHEJ). Detectable as a chromatin modification on H2AK15ub, pUbT12 accumulates in nuclear foci and is increased upon DNA damage. Mutating Thr12 prevents the removal of ubiquitin from H2AK15ub by USP51 deubiquitinating enzyme, leading to a pronounced accumulation of ubiquitinated chromatin. Chromatin modified by pUbT12 is inaccessible to 53BP1 but permissive to the homologous recombination (HR) proteins RNF169, RAD51, and the BRCA1/BARD1 complex. Phosphorylation of ubiquitin at Thr12 in the chromatin context is a new histone mark, H2AK15pUbT12, that regulates the DDR by hampering the activity of 53BP1 at damaged chromosomes.
Collapse
Affiliation(s)
- Franziska Walser
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands; Oncode Institute and Department of Cell and Chemical Biology, Chemical Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Benoît Bragantini
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sibylle Burger
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Tatiana Gubser
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | - Marco Gatti
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
| | | | - Alessandra Villa
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zurich), 8093 Zurich, Switzerland
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zurich), 8093 Zurich, Switzerland
| | - Huib Ovaa
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands; Oncode Institute and Department of Cell and Chemical Biology, Chemical Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Georges Mer
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Lorenza Penengo
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
38
|
Sebastian R, Hosogane EK, Sun EG, Tran AD, Reinhold WC, Burkett S, Sturgill DM, Gudla PR, Pommier Y, Aladjem MI, Oberdoerffer P. Epigenetic Regulation of DNA Repair Pathway Choice by MacroH2A1 Splice Variants Ensures Genome Stability. Mol Cell 2020; 79:836-845.e7. [PMID: 32649884 PMCID: PMC7483679 DOI: 10.1016/j.molcel.2020.06.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/24/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
The inactive X chromosome (Xi) is inherently susceptible to genomic aberrations. Replication stress (RS) has been proposed as an underlying cause, but the mechanisms that protect from Xi instability remain unknown. Here, we show that macroH2A1.2, an RS-protective histone variant enriched on the Xi, is required for Xi integrity and female survival. Mechanistically, macroH2A1.2 counteracts its structurally distinct and equally Xi-enriched alternative splice variant, macroH2A1.1. Comparative proteomics identified a role for macroH2A1.1 in alternative end joining (alt-EJ), which accounts for Xi anaphase defects in the absence of macroH2A1.2. Genomic instability was rescued by simultaneous depletion of macroH2A1.1 or alt-EJ factors, and mice deficient for both macroH2A1 variants harbor no overt female defects. Notably, macroH2A1 splice variant imbalance affected alt-EJ capacity also in tumor cells. Together, these findings identify macroH2A1 splicing as a modulator of genome maintenance that ensures Xi integrity and may, more broadly, predict DNA repair outcome in malignant cells.
Collapse
Affiliation(s)
- Robin Sebastian
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | - Eri K Hosogane
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Eric G Sun
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Andy D Tran
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - William C Reinhold
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sandra Burkett
- Molecular Cytogenetics Core Facility, National Cancer Institute, Frederick, MD 21702, USA
| | - David M Sturgill
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Prabhakar R Gudla
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Philipp Oberdoerffer
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
39
|
Villoria MT, Gutiérrez-Escribano P, Alonso-Rodríguez E, Ramos F, Merino E, Campos A, Montoya A, Kramer H, Aragón L, Clemente-Blanco A. PP4 phosphatase cooperates in recombinational DNA repair by enhancing double-strand break end resection. Nucleic Acids Res 2020; 47:10706-10727. [PMID: 31544936 PMCID: PMC6846210 DOI: 10.1093/nar/gkz794] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/30/2019] [Accepted: 09/11/2019] [Indexed: 12/30/2022] Open
Abstract
The role of Rad53 in response to a DNA lesion is central for the accurate orchestration of the DNA damage response. Rad53 activation relies on its phosphorylation by Mec1 and its own autophosphorylation in a manner dependent on the adaptor Rad9. While the mechanism behind Rad53 activation has been well documented, less is known about the processes that counteract its activity along the repair of a DNA adduct. Here, we describe that PP4 phosphatase is required to avoid Rad53 hyper-phosphorylation during the repair of a double-strand break, a process that impacts on the phosphorylation status of multiple factors involved in the DNA damage response. PP4-dependent Rad53 dephosphorylation stimulates DNA end resection by relieving the negative effect that Rad9 exerts over the Sgs1/Dna2 exonuclease complex. Consequently, elimination of PP4 activity affects resection and repair by single-strand annealing, defects that are bypassed by reducing Rad53 hyperphosphorylation. These results confirm that Rad53 phosphorylation is controlled by PP4 during the repair of a DNA lesion and demonstrate that the attenuation of its kinase activity during the initial steps of the repair process is essential to efficiently enhance recombinational DNA repair pathways that depend on long-range resection for their success.
Collapse
Affiliation(s)
- 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
| | - Pilar Gutiérrez-Escribano
- Cell Cycle Group. Medical Research Council, London Institute of Medical Science, Du Cane Road, London W12 0NN, UK
| | - 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
| | - 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
| | - Eva Merino
- 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
| | - Adrián Campos
- 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
| | - Alex Montoya
- Biological Mass Spectrometry and Proteomics Laboratory, Medical Research Council, London Institute of Medical Science, Du Cane Road, London W12 0NN, UK
| | - Holger Kramer
- Biological Mass Spectrometry and Proteomics Laboratory, Medical Research Council, London Institute of Medical Science, Du Cane Road, London W12 0NN, UK
| | - Luis Aragón
- Cell Cycle Group. Medical Research Council, London Institute of Medical Science, Du Cane Road, London W12 0NN, UK
| | - 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
|
40
|
Gomez Godinez V, Kabbara S, Sherman A, Wu T, Cohen S, Kong X, Maravillas-Montero JL, Shi Z, Preece D, Yokomori K, Berns MW. DNA damage induced during mitosis undergoes DNA repair synthesis. PLoS One 2020; 15:e0227849. [PMID: 32343690 PMCID: PMC7188217 DOI: 10.1371/journal.pone.0227849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/01/2020] [Indexed: 12/13/2022] Open
Abstract
Understanding the mitotic DNA damage response (DDR) is critical to our comprehension of cancer, premature aging and developmental disorders which are marked by DNA repair deficiencies. In this study we use a micro-focused laser to induce DNA damage in selected mitotic chromosomes to study the subsequent repair response. Our findings demonstrate that (1) mitotic cells are capable of DNA repair as evidenced by DNA synthesis at damage sites, (2) Repair is attenuated when DNA-PKcs and ATM are simultaneously compromised, (3) Laser damage may permit the observation of previously undetected DDR proteins when damage is elicited by other methods in mitosis, and (4) Twenty five percent of mitotic DNA-damaged cells undergo a subsequent mitosis. Together these findings suggest that mitotic DDR is more complex than previously thought and may involve factors from multiple repair pathways that are better understood in interphase.
Collapse
Affiliation(s)
- Veronica Gomez Godinez
- Institute of Engineering in Medicine, University of California-San Diego, San Diego, California, United States of America
| | - Sami Kabbara
- Department of Developmental and Cell Biology, University of California-Irvine, Irvine, California, United States of America
- Beckman Laser Institute, University of California-Irvine, Irvine, California, United States of America
| | - Adria Sherman
- Institute of Engineering in Medicine, University of California-San Diego, San Diego, California, United States of America
- Beckman Laser Institute, University of California-Irvine, Irvine, California, United States of America
| | - Tao Wu
- Beckman Laser Institute, University of California-Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, California, United States of America
| | - Shirli Cohen
- Institute of Engineering in Medicine, University of California-San Diego, San Diego, California, United States of America
| | - Xiangduo Kong
- Department of Biological Chemistry, University of California-Irvine, Irvine, California, United States of America
| | | | - Zhixia Shi
- Institute of Engineering in Medicine, University of California-San Diego, San Diego, California, United States of America
| | - Daryl Preece
- Beckman Laser Institute, University of California-Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, California, United States of America
| | - Kyoko Yokomori
- Department of Biological Chemistry, University of California-Irvine, Irvine, California, United States of America
| | - Michael W. Berns
- Institute of Engineering in Medicine, University of California-San Diego, San Diego, California, United States of America
- Department of Developmental and Cell Biology, University of California-Irvine, Irvine, California, United States of America
- Beckman Laser Institute, University of California-Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California-Irvine, Irvine, California, United States of America
| |
Collapse
|
41
|
Blackford AN, Stucki M. How Cells Respond to DNA Breaks in Mitosis. Trends Biochem Sci 2020; 45:321-331. [PMID: 32001093 DOI: 10.1016/j.tibs.2019.12.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/05/2019] [Accepted: 12/31/2019] [Indexed: 12/16/2022]
Abstract
DNA double-strand breaks (DSBs) are highly toxic lesions that can lead to chromosomal instability if they are not repaired correctly. DSBs are especially dangerous in mitosis when cells go through the complex process of equal chromosome segregation into daughter cells. When cells encounter DSBs in interphase, they are able to arrest the cell cycle until the breaks are repaired before entering mitosis. However, when DSBs occur during mitosis, cells no longer arrest but prioritize completion of cell division over repair of DNA damage. This review focuses on recent progress in our understanding of the mechanisms that allow mitotic cells to postpone DSB repair without accumulating massive chromosomal instability. Additionally, we review possible physiological consequences of failed DSB responses in mitosis.
Collapse
Affiliation(s)
- Andrew N Blackford
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK; Cancer Research UK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK.
| | - Manuel Stucki
- Department of Gynecology, University of Zurich, Wagistrasse 14, CH-8952 Schlieren, Switzerland.
| |
Collapse
|
42
|
Wang B, Zhu XX, Pan LY, Chen HF, Shen XY. PP4C facilitates lung cancer proliferation and inhibits apoptosis via activating MAPK/ERK pathway. Pathol Res Pract 2020; 216:152910. [PMID: 32139257 DOI: 10.1016/j.prp.2020.152910] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/09/2020] [Accepted: 02/28/2020] [Indexed: 12/26/2022]
Abstract
PURPOSE Protein phosphatase 4 catalytic subunit (PP4C) has been shown to play crucial regulatory roles in biological process and is frequently upregulated in cancer such as breast and colorectal carcinoma. However, the function and potential molecular mechanism of PP4C in lung cancer remains unclear. METHODS Bioinformatic analysis was used to detect the expression level and prognosis of patients. Western blot, quantitative real-time PCR (qRT-PCR), CCK8, 5-Ethynyl-2'-deoxyuridine (Edu) proliferation assay and flow cytometric were used to explore the function in lung cancer cells. RESULTS In this study, we found that PP4C was upregulated in lung cancer tissues as compared with that in normal lung tissues. Furthermore, patients with high expression level of PP4C were correlated with a poor prognosis in lung cancer patients. In vitro, CCK8, Edu proliferation assays and flow cytometry analysis showed that PP4C could promote lung cancer cell growth and inhibit apoptosis. Mechanistic investigations revealed that PP4C may interact with PP4R1 and promote ERK activation. Additionally, PP4C depletion resulted in lower tumor growth in vivo. CONCLUSIONS Taken together, these data showed the oncogenic of PP4C in NSCLC tumorigenesis and provide a new insight of PP4C in the progression of NSCLC.
Collapse
Affiliation(s)
- Bin Wang
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, No. 221 West Yan-an Road, Shanghai 200040, China
| | - Xun-Xia Zhu
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, No. 221 West Yan-an Road, Shanghai 200040, China
| | - Lin-Yue Pan
- Department of Respiration, The Affiliated Huadong Hospital of Fudan University, No. 221 West Yan-an Road, Shanghai 200040, China
| | - He-Feng Chen
- Department of Respiration, The Affiliated Huadong Hospital of Fudan University, No. 221 West Yan-an Road, Shanghai 200040, China.
| | - Xiao-Yong Shen
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, No. 221 West Yan-an Road, Shanghai 200040, China.
| |
Collapse
|
43
|
Wang B, Pan LY, Kang N, Shen XY. PP4R1 interacts with HMGA2 to promote non-small-cell lung cancer migration and metastasis via activating MAPK/ERK-induced epithelial-mesenchymal transition. Mol Carcinog 2020; 59:467-477. [PMID: 32077156 DOI: 10.1002/mc.23168] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/26/2020] [Accepted: 02/06/2020] [Indexed: 12/21/2022]
Abstract
Protein phosphatase 4 regulatory subunit 1 (PP4R1) has been shown to play a role in the regulation of centrosome maturation, apoptosis, DNA repair, and tumor necrosis factor signaling. However, the function of PP4R1 in non-small-cell lung cancer remains unclear. In this study, we identify PP4R1 as an oncogene through Oncomine database mining and immunohistochemical staining, and we showed that PP4R1 is upregulated in lung cancer tissues as compared with that in normal lung tissues and correlated with a poor prognosis in lung cancer patients. Furthermore, in vitro study by wound-healing and Transwell assay showed that PP4R1 could promote migration and invasion of lung cancer cells. Mechanistic investigations revealed that PP4R1 could cooperate with high mobility group AT-hook 2 and thereby promotes epithelial-mesenchymal transition via MAPK/extracellular receptor kinase activation. Taken together, our study provides a rich resource for understanding PP4R1 in lung cancer and indicates that PP4R1 may serve as a potential biomarker in lung cancer therapies.
Collapse
Affiliation(s)
- Bin Wang
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Lin-Yue Pan
- Department of Respiration, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Ning Kang
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Xiao-Yong Shen
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| |
Collapse
|
44
|
Entrenching role of cell cycle checkpoints and autophagy for maintenance of genomic integrity. DNA Repair (Amst) 2019; 86:102748. [PMID: 31790874 DOI: 10.1016/j.dnarep.2019.102748] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/19/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022]
Abstract
Genomic integrity of the cell is crucial for the successful transmission of genetic information to the offspring and its survival. Persistent DNA damage induced by endogenous and exogenous agents leads to various metabolic manifestations. To combat this, eukaryotes have developed complex DNA damage response (DDR) pathway which senses the DNA damage and activates an arsenal of enzymes for the repair of damaged DNA. The active pathways for DNA repair are nucleotide excision repair (NER), base excision repair (BER) and mismatch repair (MMR) for single-strand break repair whereas homologous recombination (HR) and non-homologous end-joining (NHEJ) for double-strand break repair. OGG1 is a DNA glycosylase which initiates BER while Mre11-Rad50-Nbs1 (MRN) protein complex is the primary responder to DSBs which gets localized to damage sites. DNA damage response is meticulously executed by three related kinases: ATM, ATR, and DNA-PK. ATM- and ATR-dependent phosphorylation of p53, Chk1, and Chk2 regulate the G1/S, intra-S, or G2/M checkpoints of the cell cycle, respectively. Autophagy is an evolutionarily conserved process that plays a pivotal role in the regulation of DNA repair and maintains the cellular homeostasis. Genotoxic stress-induced altered autophagy occurs in a P53 dependent manner which is also the master regulator of genotoxic stress. A plethora of proteins involved in autophagy is regulated by p53 which involve DRAM, DAPK, and AMPK. As evident, the mtDNA is more prone to damage than nuclear DNA because of its close proximity to the site of ROS generation. Depending on the extent of damage either the repair mechanism or mitophagy gets triggered. SIRT1 is the master regulator which directs the stress response to mitophagy. Nix, a LC3 adapter also participates in Parkin mediated mitophagy. This review highlights the intricate crosstalks between DNA damage and cell cycle checkpoints activation. The DNA damage mediated regulation of autophagy and mitophagy is also reviewed in detail.
Collapse
|
45
|
Scully R, Panday A, Elango R, Willis NA. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat Rev Mol Cell Biol 2019; 20:698-714. [PMID: 31263220 PMCID: PMC7315405 DOI: 10.1038/s41580-019-0152-0] [Citation(s) in RCA: 951] [Impact Index Per Article: 158.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2019] [Indexed: 11/09/2022]
Abstract
The major pathways of DNA double-strand break (DSB) repair are crucial for maintaining genomic stability. However, if deployed in an inappropriate cellular context, these same repair functions can mediate chromosome rearrangements that underlie various human diseases, ranging from developmental disorders to cancer. The two major mechanisms of DSB repair in mammalian cells are non-homologous end joining (NHEJ) and homologous recombination. In this Review, we consider DSB repair-pathway choice in somatic mammalian cells as a series of 'decision trees', and explore how defective pathway choice can lead to genomic instability. Stalled, collapsed or broken DNA replication forks present a distinctive challenge to the DSB repair system. Emerging evidence suggests that the 'rules' governing repair-pathway choice at stalled replication forks differ from those at replication-independent DSBs.
Collapse
Affiliation(s)
- Ralph Scully
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Arvind Panday
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Rajula Elango
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Nicholas A Willis
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Parmar K, Kochupurakkal BS, Lazaro JB, Wang ZC, Palakurthi S, Kirschmeier PT, Yang C, Sambel LA, Farkkila A, Reznichenko E, Reavis HD, Dunn CE, Zou L, Do KT, Konstantinopoulos PA, Matulonis UA, Liu JF, D’Andrea AD, Shapiro GI. The CHK1 Inhibitor Prexasertib Exhibits Monotherapy Activity in High-Grade Serous Ovarian Cancer Models and Sensitizes to PARP Inhibition. Clin Cancer Res 2019; 25:6127-6140. [PMID: 31409614 PMCID: PMC6801076 DOI: 10.1158/1078-0432.ccr-19-0448] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/24/2019] [Accepted: 07/16/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE PARP inhibitors are approved for the treatment of high-grade serous ovarian cancers (HGSOC). Therapeutic resistance, resulting from restoration of homologous recombination (HR) repair or replication fork stabilization, is a pressing clinical problem. We assessed the activity of prexasertib, a checkpoint kinase 1 (CHK1) inhibitor known to cause replication catastrophe, as monotherapy and in combination with the PARP inhibitor olaparib in preclinical models of HGSOC, including those with acquired PARP inhibitor resistance. EXPERIMENTAL DESIGN Prexasertib was tested as a single agent or in combination with olaparib in 14 clinically annotated and molecularly characterized luciferized HGSOC patient-derived xenograft (PDX) models and in a panel of ovarian cancer cell lines. The ability of prexasertib to impair HR repair and replication fork stability was also assessed. RESULTS Prexasertib monotherapy demonstrated antitumor activity across the 14 PDX models. Thirteen models were resistant to olaparib monotherapy, including 4 carrying BRCA1 mutation. The combination of olaparib with prexasertib was synergistic and produced significant tumor growth inhibition in an olaparib-resistant model and further augmented the degree and durability of response in the olaparib-sensitive model. HGSOC cell lines, including those with acquired PARP inhibitor resistance, were also sensitive to prexasertib, associated with induction of DNA damage and replication stress. Prexasertib also sensitized these cell lines to PARP inhibition and compromised both HR repair and replication fork stability. CONCLUSIONS Prexasertib exhibits monotherapy activity in PARP inhibitor-resistant HGSOC PDX and cell line models, reverses restored HR and replication fork stability, and synergizes with PARP inhibition.
Collapse
Affiliation(s)
- Kalindi Parmar
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bose S. Kochupurakkal
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jean-Bernard Lazaro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhigang C. Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sangeetha Palakurthi
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Paul T. Kirschmeier
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Chunyu Yang
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Larissa A. Sambel
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anniina Farkkila
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elizaveta Reznichenko
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hunter D Reavis
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Connor E. Dunn
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lee Zou
- Department of Pathology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts
| | - Khanh T. Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Panagiotis A. Konstantinopoulos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ursula A. Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joyce F. Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Alan D. D’Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Geoffrey I. Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts,Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Krenning L, van den Berg J, Medema RH. Life or Death after a Break: What Determines the Choice? Mol Cell 2019; 76:346-358. [PMID: 31561953 DOI: 10.1016/j.molcel.2019.08.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 01/22/2023]
Abstract
DNA double-strand breaks (DSBs) pose a constant threat to genomic integrity. Such DSBs need to be repaired to preserve homeostasis at both the cellular and organismal levels. Hence, the DNA damage response (DDR) has evolved to repair these lesions and limit their toxicity. The initiation of DNA repair depends on the activation of the DDR, and we know that the strength of DDR signaling may differentially affect cellular viability. However, we do not fully understand what determines the cytotoxicity of a DSB. Recent work has identified genomic location, (in)correct DNA repair pathway usage, and cell-cycle position as contributors to DSB-induced cytotoxicity. In this review, we discuss how these determinants affect cytotoxicity, highlight recent discoveries, and identify open questions that could help to improve our understanding about cell fate decisions after a DNA DSB.
Collapse
Affiliation(s)
- Lenno Krenning
- Division of Cell Biology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jeroen van den Berg
- Division of Cell Biology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - René H Medema
- Division of Cell Biology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
48
|
Zheng XF, Acharya SS, Choe KN, Nikhil K, Adelmant G, Satapathy SR, Sharma S, Viccaro K, Rana S, Natarajan A, Sicinski P, Marto JA, Shah K, Chowdhury D. A mitotic CDK5-PP4 phospho-signaling cascade primes 53BP1 for DNA repair in G1. Nat Commun 2019; 10:4252. [PMID: 31534152 PMCID: PMC6751209 DOI: 10.1038/s41467-019-12084-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/20/2019] [Indexed: 01/10/2023] Open
Abstract
Mitotic cells attenuate the DNA damage response (DDR) by phosphorylating 53BP1, a critical DDR mediator, to prevent its localization to damaged chromatin. Timely dephosphorylation of 53BP1 is critical for genome integrity, as premature recruitment of 53BP1 to DNA lesions impairs mitotic fidelity. Protein phosphatase 4 (PP4) dephosphorylates 53BP1 in late mitosis to allow its recruitment to DNA lesions in G1. How cells appropriately dephosphorylate 53BP1, thereby restoring DDR, is unclear. Here, we elucidate the underlying mechanism of kinetic control of 53BP1 dephosphorylation in mitosis. We demonstrate that CDK5, a kinase primarily functional in post-mitotic neurons, is active in late mitotic phases in non-neuronal cells and directly phosphorylates PP4R3β, the PP4 regulatory subunit that recognizes 53BP1. Specific inhibition of CDK5 in mitosis abrogates PP4R3β phosphorylation and abolishes its recognition and dephosphorylation of 53BP1, ultimately preventing the localization of 53BP1 to damaged chromatin. Our results establish CDK5 as a regulator of 53BP1 recruitment.
Collapse
Affiliation(s)
- Xiao-Feng Zheng
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Sanket S Acharya
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Katherine N Choe
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Kumar Nikhil
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Guillaume Adelmant
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shakti Ranjan Satapathy
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Samanta Sharma
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Keith Viccaro
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Sandeep Rana
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Peter Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Dipanjan Chowdhury
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
49
|
Burger K, Ketley RF, Gullerova M. Beyond the Trinity of ATM, ATR, and DNA-PK: Multiple Kinases Shape the DNA Damage Response in Concert With RNA Metabolism. Front Mol Biosci 2019; 6:61. [PMID: 31428617 PMCID: PMC6688092 DOI: 10.3389/fmolb.2019.00061] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/11/2019] [Indexed: 12/22/2022] Open
Abstract
Our genome is constantly exposed to endogenous and exogenous sources of DNA damage resulting in various alterations of the genetic code. DNA double-strand breaks (DSBs) are considered one of the most cytotoxic lesions. Several types of repair pathways act to repair DNA damage and maintain genome stability. In the canonical DNA damage response (DDR) DSBs are recognized by the sensing kinases Ataxia-telangiectasia mutated (ATM), Ataxia-telangiectasia and Rad3-related (ATR), and DNA-dependent protein kinase (DNA-PK), which initiate a cascade of kinase-dependent amplification steps known as DSB signaling. Recent evidence suggests that efficient recognition and repair of DSBs relies on the transcription and processing of non-coding (nc)RNA molecules by RNA polymerase II (RNAPII) and the RNA interference (RNAi) factors Drosha and Dicer. Multiple kinases influence the phosphorylation status of both the RNAPII carboxy-terminal domain (CTD) and Dicer in order to regulate RNA-dependent DSBs repair. The importance of kinase signaling and RNA processing in the DDR is highlighted by the regulation of p53-binding protein (53BP1), a key regulator of DSB repair pathway choice between homologous recombination (HR) and non-homologous end joining (NHEJ). Additionally, emerging evidence suggests that RNA metabolic enzymes also play a role in the repair of other types of DNA damage, including the DDR to ultraviolet radiation (UVR). RNAi factors are also substrates for mitogen-activated protein kinase (MAPK) signaling and mediate the turnover of ncRNA during nucleotide excision repair (NER) in response to UVR. Here, we review kinase-dependent phosphorylation events on RNAPII, Drosha and Dicer, and 53BP1 that modulate the key steps of the DDR to DSBs and UVR, suggesting an intimate link between the DDR and RNA metabolism.
Collapse
Affiliation(s)
| | | | - Monika Gullerova
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Interplay between Phosphatases and the Anaphase-Promoting Complex/Cyclosome in Mitosis. Cells 2019; 8:cells8080814. [PMID: 31382469 PMCID: PMC6721574 DOI: 10.3390/cells8080814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Accurate division of cells into two daughters is a process that is vital to propagation of life. Protein phosphorylation and selective degradation have emerged as two important mechanisms safeguarding the delicate choreography of mitosis. Protein phosphatases catalyze dephosphorylation of thousands of sites on proteins, steering the cells through establishment of the mitotic phase and exit from it. A large E3 ubiquitin ligase, the anaphase-promoting complex/cyclosome (APC/C) becomes active during latter stages of mitosis through G1 and marks hundreds of proteins for destruction. Recent studies have revealed the complex interregulation between these two classes of enzymes. In this review, we highlight the direct and indirect mechanisms by which phosphatases and the APC/C mutually influence each other to ensure accurate spatiotemporal and orderly progression through mitosis, with a particular focus on recent insights and conceptual advances.
Collapse
|