1
|
Barbier MT, Del Valle L. Co-Detection of EBV and Human Polyomavirus JCPyV in a Case of AIDS-Related Multifocal Primary Central Nervous System Diffuse Large B-Cell Lymphoma. Viruses 2023; 15:755. [PMID: 36992464 PMCID: PMC10059075 DOI: 10.3390/v15030755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
The human neurotropic Polyomavirus JCPyV is the widespread opportunistic causative pathogen of the fatal demyelinating disease progressive multifocal leukoencephalopathy; however, it has also been implicated in the oncogenesis of several types of cancers. It causes brain tumors when intracerebrally inoculated into rodents, and genomic sequences of different strains and expression of the viral protein large T-Antigen have been detected in a wide variety of glial brain tumors and CNS lymphomas. Here, we present a case of an AIDS-related multifocal primary CNS lymphoma in which JCPyV genomic sequences of the three regions of JCPyV and expression of T-Antigen were detected by PCR and immunohistochemistry, respectively. No capsid proteins were detected, ruling out active JCPyV replication. Sequencing of the control region revealed that Mad-4 was the strain of JCPyV present in tumor cells. In addition, expression of viral proteins LMP and EBNA-1 from another ubiquitous oncogenic virus, Epstein-Barr, was also detected in the same lymphocytic neoplastic cells, co-localizing with JCPyV T-Antigen, suggesting a potential collaboration between these two viruses in the process of malignant transformation of B-lymphocytes, which are the site of latency and reactivation for both viruses.
Collapse
Affiliation(s)
- Mallory T. Barbier
- Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Luis Del Valle
- Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Pathology, Louisiana State University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
2
|
Molecular Alterations and Putative Therapeutic Targeting of Planar Cell Polarity Proteins in Breast Cancer. J Clin Med 2023; 12:jcm12020411. [PMID: 36675340 PMCID: PMC9864096 DOI: 10.3390/jcm12020411] [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: 11/23/2022] [Revised: 12/27/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Treatment and outcomes of breast cancer, one of the most prevalent female cancers, have improved in recent decades. However, metastatic breast cancer remains incurable in most cases, and new therapies are needed to ameliorate prognosis. Planar cell polarity (PCP) is a characteristic of epithelial cells that form layers and is integral to the communication of these cells with neighboring cells. Dysfunction of PCP is observed in cancers and may confer a targetable vulnerability. METHODS The breast cancer cohorts from The Cancer Genome Atlas (TCGA) and the METABRIC study were interrogated for molecular alterations in genes of the PCP pathway. The groups with the most prevalent alterations were characterized, and survival was compared with counterparts not possessing PCP alterations. Breast cancer cell lines with PCP alterations from the Cancer Cell Line Encyclopedia (CCLE) were interrogated for sensitivity to drugs affecting PCP. RESULTS Among genes of the PCP pathway, VANGL2, NOS1AP and SCRIB display amplifications in a sizable minority of breast cancers. Concomitant up-regulation at the mRNA level can be observed mostly in basal cancers, but it does not correlate well with the amplification status of the genes, as it can also be observed in non-amplified cases. In an exploration of cell line models, two of the four breast cancer cell line models with amplifications in VANGL2, NOS1AP and SCRIB display sensitivity to drugs inhibiting acyl-transferase porcupine interfering with the WNT pathway. This sensitivity suggests a possible therapeutic role of these inhibitors in cancers bearing the amplifications. CONCLUSION Molecular alterations in PCP genes can be observed in breast cancers with a predilection for the basal sub-type. An imperfect correlation of copy number alterations with mRNA expression suggests that post-translational modifications are important in PCP regulation. Inhibitors of acyl-transferase porcupine may be rational candidates for combination therapy development in PCP-altered breast cancers.
Collapse
|
3
|
Muoio D, Laspata N, Fouquerel E. Functions of ADP-ribose transferases in the maintenance of telomere integrity. Cell Mol Life Sci 2022; 79:215. [PMID: 35348914 PMCID: PMC8964661 DOI: 10.1007/s00018-022-04235-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022]
Abstract
The ADP-ribose transferase (ART) family comprises 17 enzymes that catalyze mono- or poly-ADP-ribosylation, a post-translational modification of proteins. Present in all subcellular compartments, ARTs are implicated in a growing number of biological processes including DNA repair, replication, transcription regulation, intra- and extra-cellular signaling, viral infection and cell death. Five members of the family, PARP1, PARP2, PARP3, tankyrase 1 and tankyrase 2 are mainly described for their crucial functions in the maintenance of genome stability. It is well established that the most describedrole of PARP1, 2 and 3 is the repair of DNA lesions while tankyrases 1 and 2 are crucial for maintaining the integrity of telomeres. Telomeres, nucleoprotein complexes located at the ends of eukaryotic chromosomes, utilize their unique structure and associated set of proteins to orchestrate the mechanisms necessary for their own protection and replication. While the functions of tankyrases 1 and 2 at telomeres are well known, several studies have also brought PARP1, 2 and 3 to the forefront of telomere protection. The singular quality of the telomeric environment has highlighted protein interactions and molecular pathways distinct from those described throughout the genome. The aim of this review is to provide an overview of the current knowledge on the multiple roles of PARP1, PARP2, PARP3, tankyrase 1 and tankyrase 2 in the maintenance and preservation of telomere integrity.
Collapse
Affiliation(s)
- Daniela Muoio
- UPMC Cancer Institute and Department of Pharmacology and Chemical Biology at the University of Pittsburgh, Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Natalie Laspata
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 S. 10th street, Philadelphia, PA, 19107, USA
| | - Elise Fouquerel
- UPMC Cancer Institute and Department of Pharmacology and Chemical Biology at the University of Pittsburgh, Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
4
|
Yu M, Yang Y, Sykes M, Wang S. Small-Molecule Inhibitors of Tankyrases as Prospective Therapeutics for Cancer. J Med Chem 2022; 65:5244-5273. [PMID: 35306814 DOI: 10.1021/acs.jmedchem.1c02139] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tankyrases are multifunctional poly(adenosine diphosphate-ribose) polymerases that regulate diverse biological processes including telomere maintenance and cellular signaling. These processes are often implicated in a number of human diseases, with cancer being the most prevalent example. Accordingly, tankyrase inhibitors have gained increasing attention as potential therapeutics. Since the discovery of XAV939 and IWR-1 as the first tankyrase inhibitors over two decades ago, tankyrase-targeted drug discovery has made significant progress. This review starts with an introduction of tankyrases, with emphasis placed on their cancer-related functions. Small-molecule inhibitors of tankyrases are subsequently delineated based on their distinct modes of binding to the enzymes. In addition to inhibitors that compete with oxidized nicotinamide adenine dinucleotide (NAD+) for binding to the catalytic domain of tankyrases, non-NAD+-competitive inhibitors are detailed. This is followed by a description of three clinically trialled tankyrase inhibitors. To conclude, some of challenges and prospects in developing tankyrase-targeted cancer therapies are discussed.
Collapse
Affiliation(s)
- Mingfeng Yu
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Yuchao Yang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Matthew Sykes
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Shudong Wang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
5
|
Vilchez Larrea S, Valsecchi WM, Fernández Villamil SH, Lafon Hughes LI. First body of evidence suggesting a role of a tankyrase-binding motif (TBM) of vinculin (VCL) in epithelial cells. PeerJ 2021; 9:e11442. [PMID: 34123588 PMCID: PMC8164839 DOI: 10.7717/peerj.11442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Adherens junctions (AJ) are involved in cancer, infections and neurodegeneration. Still, their composition has not been completely disclosed. Poly(ADP-ribose) polymerases (PARPs) catalyze the synthesis of poly(ADP-ribose) (PAR) as a posttranslational modification. Four PARPs synthesize PAR, namely PARP-1/2 and Tankyrase-1/2 (TNKS). In the epithelial belt, AJ are accompanied by a PAR belt and a subcortical F-actin ring. F-actin depolymerization alters the AJ and PAR belts while PARP inhibitors prevent the assembly of the AJ belt and cortical actin. We wondered which PARP synthesizes the belt and which is the PARylation target protein. Vinculin (VCL) participates in the anchorage of F-actin to the AJ, regulating its functions, and colocalized with the PAR belt. TNKS has been formerly involved in the assembly of epithelial cell junctions. HYPOTHESIS TNKS poly(ADP-ribosylates) (PARylates) epithelial belt VCL, affecting its functions in AJ, including cell shape maintenance. MATERIALS AND METHODS Tankyrase-binding motif (TBM) sequences in hVCL gene were identified and VCL sequences from various vertebrates, Drosophila melanogaster and Caenorhabditis elegans were aligned and compared. Plasma membrane-associated PAR was tested by immunocytofluorescence (ICF) and subcellular fractionation in Vero cells while TNKS role in this structure and cell junction assembly was evaluated using specific inhibitors. The identity of the PARylated proteins was tested by affinity precipitation with PAR-binding reagent followed by western blots. Finally, MCF-7 human breast cancer epithelial cells were subjected to transfection with Tol2-plasmids, carrying a dicistronic expression sequence including Gallus gallus wt VCL (Tol-2-GgVCL), or the same VCL gene with a point mutation in TBM-II (Tol2-GgVCL/*TBM) under the control of a β-actin promoter, plus green fluorescent protein following an internal ribosome entry site (IRES-GFP) to allow the identification of transfected cells without modifying the transfected protein of interest. RESULTS AND DISCUSSION In this work, some of the hypothesis predictions have been tested. We have demonstrated that: (1) VCL TBMs were conserved in vertebrate evolution while absent in C. elegans; (2) TNKS inhibitors disrupted the PAR belt synthesis, while PAR and an endogenous TNKS pool were associated to the plasma membrane; (3) a VCL pool was covalently PARylated; (4) transfection of MCF-7 cells leading to overexpression of Gg-VCL/*TBM induced mesenchymal-like cell shape changes. This last point deserves further investigation, bypassing the limits of our transient transfection and overexpression system. In fact, a 5th testable prediction would be that a single point mutation in VCL TBM-II under endogenous expression control would induce an epithelial to mesenchymal transition (EMT). To check this, a CRISPR/Cas9 substitution approach followed by migration, invasion, gene expression and chemo-resistance assays should be performed.
Collapse
Affiliation(s)
- Salomé Vilchez Larrea
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Ciudad Autónoma de Buenos Aires, República Argentina
| | - Wanda Mariela Valsecchi
- Instituto de Química y Fisicoquímica Biológicas, “Prof. Alejandro C. Paladini” (IQUIFIB) Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Ciudad Autónoma de Buenos Aires, República Argentina
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvia H. Fernández Villamil
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Ciudad Autónoma de Buenos Aires, República Argentina
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Laura I. Lafon Hughes
- Grupo de Biofisicoquímica, Departamento de Ciencias Biológicas, Centro Universitario Regional Litoral Norte (CENUR), Universidad de la República, Salto, Uruguay
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Ministerio de Educación y Cultura, Montevideo, Uruguay
| |
Collapse
|
6
|
Abstract
Effective maintenance and stability of our genomes is essential for normal cell division, tissue homeostasis, and cellular and organismal fitness. The processes of chromosome replication and segregation require continual surveillance to insure fidelity. Accurate and efficient repair of DNA damage preserves genome integrity, which if lost can lead to multiple diseases, including cancer. Poly(ADP-ribose) a dynamic and reversible posttranslational modification and the enzymes that catalyze it (PARP1, PARP2, tankyrase 1, and tankyrase 2) function to maintain genome stability through diverse mechanisms. Here we review the role of these enzymes and the modification in genome repair, replication, and resolution in human cells.
Collapse
Affiliation(s)
- Kameron Azarm
- Department of Pathology, Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York 10016, USA
| | - Susan Smith
- Department of Pathology, Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
7
|
Eisemann T, Pascal JM. Poly(ADP-ribose) polymerase enzymes and the maintenance of genome integrity. Cell Mol Life Sci 2020; 77:19-33. [PMID: 31754726 PMCID: PMC11104942 DOI: 10.1007/s00018-019-03366-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/11/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023]
Abstract
DNA damage response (DDR) relies on swift and accurate signaling to rapidly identify DNA lesions and initiate repair. A critical DDR signaling and regulatory molecule is the posttranslational modification poly(ADP-ribose) (PAR). PAR is synthesized by a family of structurally and functionally diverse proteins called poly(ADP-ribose) polymerases (PARPs). Although PARPs share a conserved catalytic domain, unique regulatory domains of individual family members endow PARPs with unique properties and cellular functions. Family members PARP-1, PARP-2, and PARP-3 (DDR-PARPs) are catalytically activated in the presence of damaged DNA and act as damage sensors. Family members tankyrase-1 and closely related tankyrase-2 possess SAM and ankyrin repeat domains that regulate their diverse cellular functions. Recent studies have shown that the tankyrases share some overlapping functions with the DDR-PARPs, and even perform novel functions that help preserve genomic integrity. In this review, we briefly touch on DDR-PARP functions, and focus on the emerging roles of tankyrases in genome maintenance. Preservation of genomic integrity thus appears to be a common function of several PARP family members, depicting PAR as a multifaceted guardian of the genome.
Collapse
Affiliation(s)
- Travis Eisemann
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - John M Pascal
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
8
|
Abstract
Mitosis ensures accurate segregation of duplicated DNA through tight regulation of chromosome condensation, bipolar spindle assembly, chromosome alignment in the metaphase plate, chromosome segregation and cytokinesis. Poly(ADP-ribose) polymerases (PARPs), in particular PARP1, PARP2, PARP3, PARP5a (TNKS1), as well as poly(ADP-ribose) glycohydrolase (PARG), regulate different mitotic functions, including centrosome function, mitotic spindle assembly, mitotic checkpoints, telomere length and telomere cohesion. PARP depletion or inhibition give rise to various mitotic defects such as centrosome amplification, multipolar spindles, chromosome misalignment, premature loss of cohesion, metaphase arrest, anaphase DNA bridges, lagging chromosomes, and micronuclei. As the mechanisms of PARP1/2 inhibitor-mediated cell death are being progressively elucidated, it is becoming clear that mitotic defects caused by PARP1/2 inhibition arise due to replication stress and DNA damage in S phase. As it stands, entrapment of inactive PARP1/2 on DNA phenocopies replication stress through accumulation of unresolved replication intermediates, double-stranded DNA breaks (DSBs) and incorrectly repaired DSBs, which can be transmitted from S phase to mitosis and instigate various mitotic defects, giving rise to both numerical and structural chromosomal aberrations. Cancer cells have increased levels of replication stress, which makes them particularly susceptible to a combination of agents that compromise replication fork stability. Indeed, combining PARP1/2 inhibitors with genetic deficiencies in DNA repair pathways, DNA-damaging agents, ATR and other cell cycle checkpoint inhibitors has yielded synergistic effects in killing cancer cells. Here I provide a comprehensive overview of the mitotic functions of PARPs and PARG, mitotic phenotypes induced by their depletion or inhibition, as well as the therapeutic relevance of targeting mitotic cells by directly interfering with mitotic functions or indirectly through replication stress.
Collapse
Affiliation(s)
- Dea Slade
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-gasse 9, 1030 Vienna, Austria.
| |
Collapse
|
9
|
Zhou T, Yi F, Wang Z, Guo Q, Liu J, Bai N, Li X, Dong X, Ren L, Cao L, Song X. The Functions of DNA Damage Factor RNF8 in the Pathogenesis and Progression of Cancer. Int J Biol Sci 2019; 15:909-918. [PMID: 31182912 PMCID: PMC6535783 DOI: 10.7150/ijbs.31972] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/08/2019] [Indexed: 12/31/2022] Open
Abstract
The really interesting new gene (RING) finger protein 8 (RNF8) is a central factor in DNA double strand break (DSB) signal transduction. DSB damage is the most toxic type of DNA damage to cells and is related to genomic instability. Multiple roles for RNF8 have been identified in DNA damage response as well as in other functions, such as telomere protection, cell cycle control and transcriptional regulation. These functions are closely correlated to tumorigenesis and cancer progression. Indeed, deficiency of RNF8 caused spontaneous tumorigenesis in a mouse model. Deciphering these mechanisms of RNF8 may shed light on strategies for cancer treatment. In this review, we summarize the current understanding of both classical and nonclassical functions of RNF8, and discuss its roles in the pathogenesis and progression of tumor.
Collapse
Affiliation(s)
- Tingting Zhou
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Fei Yi
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Zhuo Wang
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Qiqiang Guo
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Jingwei Liu
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Ning Bai
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Xiaoman Li
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Xiang Dong
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Ling Ren
- Department of Anus and Intestine Surgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Liu Cao
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| | - Xiaoyu Song
- Institute of Translational Medicine, China Medical University; Key Laboratory of Medical Cell Biology, Ministry of Education; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning Province, China
| |
Collapse
|
10
|
Hou H, Cooper JP. Stretching, scrambling, piercing and entangling: Challenges for telomeres in mitotic and meiotic chromosome segregation. Differentiation 2018; 100:12-20. [PMID: 29413748 DOI: 10.1016/j.diff.2018.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 12/24/2022]
Abstract
The consequences of telomere loss or dysfunction become most prominent when cells enter the nuclear division stage of the cell cycle. At this climactic stage when chromosome segregation occurs, telomere fusions or entanglements can lead to chromosome breakage, wreaking havoc on genome stability. Here we review recent progress in understanding the mechanisms of detangling and breaking telomere associations at mitosis, as well as the unique ways in which telomeres are processed to allow regulated sister telomere separation. Moreover, we discuss unexpected roles for telomeres in orchestrating nuclear envelope breakdown and spindle formation, crucial processes for nuclear division. Finally, we discuss the discovery that telomeres create microdomains in the nucleus that are conducive to centromere assembly, cementing the unexpectedly influential role of telomeres in mitosis.
Collapse
Affiliation(s)
- Haitong Hou
- Telomere Biology Section, LBMB, NCI, NIH, Building 37, Room 6050, Bethesda MD 20892, USA
| | - Julia Promisel Cooper
- Telomere Biology Section, LBMB, NCI, NIH, Building 37, Room 6050, Bethesda MD 20892, USA.
| |
Collapse
|
11
|
Inhibitors of telomerase and poly(ADP-ribose) polymerases synergize to limit the lifespan of pancreatic cancer cells. Oncotarget 2017; 8:83754-83767. [PMID: 29137380 PMCID: PMC5663552 DOI: 10.18632/oncotarget.19410] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023] Open
Abstract
Imetelstat (GRN163L) is a potent and selective inhibitor of telomerase. We have previously reported that GRN163L could shorten telomeres and limit the lifespan of CD18/HPAF and CAPAN1 pancreatic cancer cells. Here, we examined the effects of GRN163L on two other pancreatic cancer cell lines: AsPC1 and L3.6pl. In both lines, chronic exposure to GRN163L led to an initial shortening of telomeres followed by a stabilization of extremely short telomeres. In AsPC1 cells, telomere attrition eventually led to the induction of crisis and the loss of the treated population. In L3.6pl cells, crisis was transient and followed by the emergence of GRN163L-resistant cells, which could grow at increasing concentrations of GRN163L. The Shelterin complex is a telomere-associated complex that limits the access of telomerase to telomeres. The telomerase inhibitory function of this complex can be enhanced by drugs that block the poly(ADP-ribosyl)ation of its TRF1 and/or TRF2 subunits. Combined treatment of the GRN163L-resistant L3.6pl cells with GRN163L and 3-aminobenzamide (3AB), a general inhibitor of poly(ADP-ribose) polymerases, led to additional telomere shortening and limited the lifespan of the resistant cells. Results from this work suggest that inhibitors of telomerase and poly(ADP-ribose) polymerases can cooperate to limit the lifespan of pancreatic cancer cells.
Collapse
|
12
|
Tripathi E, Smith S. Cell cycle-regulated ubiquitination of tankyrase 1 by RNF8 and ABRO1/BRCC36 controls the timing of sister telomere resolution. EMBO J 2016; 36:503-519. [PMID: 27993934 DOI: 10.15252/embj.201695135] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 12/19/2022] Open
Abstract
Timely resolution of sister chromatid cohesion in G2/M is essential for genome integrity. Resolution at telomeres requires the poly(ADP-ribose) polymerase tankyrase 1, but the mechanism that times its action is unknown. Here, we show that tankyrase 1 activity at telomeres is controlled by a ubiquitination/deubiquitination cycle depending on opposing ubiquitin ligase and deubiquitinase activities. In late S/G2 phase, the DNA damage-responsive E3 ligase RNF8 conjugates K63-linked ubiquitin chains to tankyrase 1, while in G1 phase such ubiquitin chains are removed by BRISC, an ABRO1/BRCC36-containing deubiquitinase complex. We show that K63-linked ubiquitin chains accumulate on tankyrase 1 in late S/G2 to promote its stabilization, association with telomeres, and resolution of cohesion. Timing of this posttranslational modification coincides with the ATM-mediated DNA damage response that occurs on functional telomeres following replication in G2. Removal of ubiquitin chains is controlled by ABRO1/BRCC36 and occurs as cells exit mitosis and enter G1, ensuring that telomere cohesion is not resolved prematurely in S phase. Our studies suggest that a cell cycle-regulated posttranslational mechanism couples resolution of telomere cohesion with completion of telomere replication to ensure genome integrity.
Collapse
Affiliation(s)
- Ekta Tripathi
- Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Susan Smith
- Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
13
|
Expanding functions of ADP-ribosylation in the maintenance of genome integrity. Semin Cell Dev Biol 2016; 63:92-101. [PMID: 27670719 DOI: 10.1016/j.semcdb.2016.09.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/19/2016] [Accepted: 09/16/2016] [Indexed: 12/21/2022]
Abstract
Cell response to genotoxic stress requires a complex network of sensors and effectors from numerous signaling and repair pathways, among them the nuclear poly(ADP-ribose) polymerase 1 (PARP1) plays a central role. PARP1 is catalytically activated in the setting of DNA breaks. It uses NAD+ as a donor and catalyses the synthesis and subsequent covalent attachment of branched ADP-ribose polymers onto itself and various acceptor proteins to promote repair. Its inhibition is now considered as an efficient therapeutic strategy to potentiate the cytotoxic effect of chemotherapy and radiation or to exploit synthetic lethality in tumours with defective homologous recombination mediated repair. Still, efforts made on understanding the role of PARylation in DNA repair continues to yield novel discoveries. Over the last years, our knowledge in this field has been particularly advanced by the discovery of novel biochemical and functional properties featuring PARP1, by the characterization of the other PARP family members and by the identification of a panel of enzymes capable of erasing poly(ADP-ribose). The aim of this review is to provide an overview of these newest findings and their relevance in genome surveillance.
Collapse
|
14
|
Nagy Z, Kalousi A, Furst A, Koch M, Fischer B, Soutoglou E. Tankyrases Promote Homologous Recombination and Check Point Activation in Response to DSBs. PLoS Genet 2016; 12:e1005791. [PMID: 26845027 PMCID: PMC4741384 DOI: 10.1371/journal.pgen.1005791] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/16/2015] [Indexed: 11/18/2022] Open
Abstract
DNA lesions are sensed by a network of proteins that trigger the DNA damage response (DDR), a signaling cascade that acts to delay cell cycle progression and initiate DNA repair. The Mediator of DNA damage Checkpoint protein 1 (MDC1) is essential for spreading of the DDR signaling on chromatin surrounding Double Strand Breaks (DSBs) by acting as a scaffold for PI3K kinases and for ubiquitin ligases. MDC1 also plays a role both in Non-Homologous End Joining (NHEJ) and Homologous Recombination (HR) repair pathways. Here we identify two novel binding partners of MDC1, the poly (ADP-ribose) Polymerases (PARPs) TNKS1 and 2. We find that TNKSs are recruited to DNA lesions by MDC1 and regulate DNA end resection and BRCA1A complex stabilization at lesions leading to efficient DSB repair by HR and proper checkpoint activation. MDC1 recruit Tankyrases to DNA lesions to regulate homologous recombination and to control check-point activation.
Collapse
Affiliation(s)
- Zita Nagy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U964, Illkirch, France
- Centre National de Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Alkmini Kalousi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U964, Illkirch, France
- Centre National de Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Audrey Furst
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U964, Illkirch, France
- Centre National de Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Marc Koch
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U964, Illkirch, France
- Centre National de Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Benoit Fischer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U964, Illkirch, France
- Centre National de Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Evi Soutoglou
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U964, Illkirch, France
- Centre National de Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
- * E-mail:
| |
Collapse
|
15
|
Cipressa F, Morciano P, Bosso G, Mannini L, Galati A, Raffa GD, Cacchione S, Musio A, Cenci G. A role for Separase in telomere protection. Nat Commun 2016; 7:10405. [PMID: 26778495 PMCID: PMC4735636 DOI: 10.1038/ncomms10405] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/08/2015] [Indexed: 12/04/2022] Open
Abstract
Drosophila telomeres are elongated by transposition of specialized retroelements rather than telomerase activity and are assembled independently of the sequence. Fly telomeres are protected by the terminin complex that localizes and functions exclusively at telomeres and by non-terminin proteins that do not serve telomere-specific functions. We show that mutations in the Drosophila Separase encoding gene Sse lead not only to endoreduplication but also telomeric fusions (TFs), suggesting a role for Sse in telomere capping. We demonstrate that Separase binds terminin proteins and HP1, and that it is enriched at telomeres. Furthermore, we show that loss of Sse strongly reduces HP1 levels, and that HP1 overexpression in Sse mutants suppresses TFs, suggesting that TFs are caused by a HP1 diminution. Finally, we find that siRNA-induced depletion of ESPL1, the Sse human orthologue, causes telomere dysfunction and HP1 level reduction in primary fibroblasts, highlighting a conserved role of Separase in telomere protection. Drosophila telomeres are elongated by transposition of specialized retroelements rather than telomerase activity. Here, the authors show that Separase is enriched at Drosophila telomeres and loss of Sse, the gene encoding Separase, leads to telomere defects, suggesting a role for Separase in telomere protection.
Collapse
Affiliation(s)
- Francesca Cipressa
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| | - Patrizia Morciano
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| | - Giuseppe Bosso
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| | - Linda Mannini
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, c/o Area di Ricerca di S. Cataldo Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Alessandra Galati
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| | - Grazia Daniela Raffa
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| | - Stefano Cacchione
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| | - Antonio Musio
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, c/o Area di Ricerca di S. Cataldo Via G. Moruzzi 1, 56124 Pisa, Italy.,Istituto Toscano Tumori, Via T. Alderotti 26N, 50139 Firenze, Italy
| | - Giovanni Cenci
- Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Viale Regina Elena 291, 00185 Rome, Italy
| |
Collapse
|
16
|
Ramamoorthy M, Smith S. Loss of ATRX Suppresses Resolution of Telomere Cohesion to Control Recombination in ALT Cancer Cells. Cancer Cell 2015; 28:357-69. [PMID: 26373281 PMCID: PMC4573400 DOI: 10.1016/j.ccell.2015.08.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/08/2015] [Accepted: 08/03/2015] [Indexed: 01/11/2023]
Abstract
The chromatin-remodeler ATRX is frequently lost in cancer cells that use ALT (alternative lengthening of telomeres) for telomere maintenance, but its function in telomere recombination is unknown. Here we show that loss of ATRX suppresses the timely resolution of sister telomere cohesion that normally occurs prior to mitosis. In the absence of ATRX, the histone variant macroH2A1.1 binds to the poly(ADP-ribose) polymerase tankyrase 1, preventing it from localizing to telomeres and resolving cohesion. The resulting persistent telomere cohesion promotes recombination between sister telomeres, while it suppresses inappropriate recombination between non-sisters. Forced resolution of sister telomere cohesion induces excessive recombination between non-homologs, genomic instability, and impaired cell growth, indicating the ATRX-macroH2A1.1-tankyrase axis as a potential therapeutic target in ALT tumors.
Collapse
Affiliation(s)
- Mahesh Ramamoorthy
- Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, Department of Pathology, NYU Langone Medical Center and School of Medicine, New York, NY 10016, USA
| | - Susan Smith
- Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, Department of Pathology, NYU Langone Medical Center and School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
17
|
Marcand S. How do telomeres and NHEJ coexist? Mol Cell Oncol 2014; 1:e963438. [PMID: 27308342 PMCID: PMC4904885 DOI: 10.4161/23723548.2014.963438] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 08/01/2014] [Accepted: 08/07/2014] [Indexed: 12/21/2022]
Abstract
The telomeres of eukaryotes are stable open double-strand ends that coexist with nonhomologous end joining (NHEJ), the repair pathway that directly ligates DNA ends generated by double-strand breaks. Since a single end-joining event between 2 telomeres generates a circular chromosome or an unstable dicentric chromosome, NHEJ must be prevented from acting on telomeres. Multiple mechanisms mediated by telomere factors act in synergy to achieve this inhibition.
Collapse
Affiliation(s)
- Stéphane Marcand
- CEA; DSV/IRCM/SIGRR/LTR; Fontenay-aux-roses; France; INSERM UMR 967; Fontenay-aux-roses; France
| |
Collapse
|
18
|
Doksani Y, de Lange T. The role of double-strand break repair pathways at functional and dysfunctional telomeres. Cold Spring Harb Perspect Biol 2014; 6:a016576. [PMID: 25228584 DOI: 10.1101/cshperspect.a016576] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Telomeres have evolved to protect the ends of linear chromosomes from the myriad of threats posed by the cellular DNA damage signaling and repair pathways. Mammalian telomeres have to block nonhomologous end joining (NHEJ), thus preventing chromosome fusions; they need to control homologous recombination (HR), which could change telomere lengths; they have to avoid activating the ATM (ataxia telangiectasia mutated) and ATR (ATM- and RAD3-related) kinase pathways, which could induce cell cycle arrest; and they have to protect chromosome ends from hyperresection. Recent studies of telomeres have provided insights into the mechanisms of NHEJ and HR, how these double-strand break (DSB) repair pathways can be thwarted, and how telomeres have co-opted DNA repair factors to help in the protection of chromosome ends. These aspects of telomere biology are reviewed here with particular emphasis on recombination, the main focus of this collection.
Collapse
Affiliation(s)
- Ylli Doksani
- Laboratory for Cell Biology and Genetics, Rockefeller University, New York, New York 10065
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, Rockefeller University, New York, New York 10065
| |
Collapse
|
19
|
Tian X, Hou W, Bai S, Fan J, Tong H, Bai Y. XAV939 promotes apoptosis in a neuroblastoma cell line via telomere shortening. Oncol Rep 2014; 32:1999-2006. [PMID: 25190315 DOI: 10.3892/or.2014.3460] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/19/2014] [Indexed: 11/05/2022] Open
Abstract
Telomeres, telomerase and tankyrase (TNKS) have an extremely important and special association with human cell aging and cancer. Telomerase activity is abnormally high in cancer cells and is accompanied by the overexpression of tankyrase 1 (TNKS1). TNKS1 is a positive regulator of telomerase activation and telomere extension in the human body, indicating that TNKS1 may be a possible therapeutic target for cancer. XAV939 is a small-molecule inhibitor of TNKS1. The objective of the present study was to investigate the apoptotic effect of XAV939 on the neuroblastoma (NB) SH-SY5Y cell line, as well as the change in telomere length and telomerase activity and elucidate the mechanism from this perspective. In the present study, we initially treated SH-SY5Y cells with XAV939 and RNA interference (RNAi)-TNKS1, and subsequently chose the optimal sequence for RNAi-TNKS1. We then measured the telomere length using quantitative real-time polymerase chain reaction (qPCR) assay, detected the telomerase activity using the ELISA kit, observed apoptotic morphology by transmission electron microscopy, and detected the percentages of apoptotic cells using flow cytometry and Hoechst 33342 staining. We also determined the invasive ability by a cell invasion assay. The results showed that short hairpin RNA-2 (shRNA-2) was the optimal sequence for RNAi-TNKS1. Treatment with both XAV939 and RNAi-TNKS1 shortened the telomere length, promoted apoptosis and reduced the invasive ability of the SH-SY5Y cells, yet had no effect on telomerase activity. XAV939 promoted apoptosis and reduced the invasiveness of SH-SY5Y cells dependent on telomere shortening, and further research should be conducted to clarify the exact mechanisms. This research may contribute to the cure of malignant NB using multi-targeted therapy with small-molecule agents.
Collapse
Affiliation(s)
- Xiaohong Tian
- Department of Tissue Engineering, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Weijian Hou
- Department of Tissue Engineering, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shuling Bai
- Department of Tissue Engineering, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jun Fan
- Department of Tissue Engineering, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hao Tong
- Department of Tissue Engineering, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yu Bai
- Department of Ophthalmology, The Second Hospital of Shijiazhuang, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
20
|
Lupo B, Trusolino L. Inhibition of poly(ADP-ribosyl)ation in cancer: old and new paradigms revisited. Biochim Biophys Acta Rev Cancer 2014; 1846:201-15. [PMID: 25026313 DOI: 10.1016/j.bbcan.2014.07.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/02/2014] [Accepted: 07/08/2014] [Indexed: 01/31/2023]
Abstract
Inhibitors of poly(ADP-ribose) polymerases actualized the biological concept of synthetic lethality in the clinical practice, yielding a paradigmatic example of translational medicine. The profound sensitivity of tumors with germline BRCA mutations to PARP1/2 blockade owes to inherent defects of the BRCA-dependent homologous recombination machinery, which are unleashed by interruption of PARP DNA repair activity and lead to DNA damage overload and cell death. Conversely, aspirant BRCA-like tumors harboring somatic DNA repair dysfunctions (a vast entity of genetic and epigenetic defects known as "BRCAness") not always align with the familial counterpart and appear not to be equally sensitive to PARP inhibition. The acquisition of secondary resistance in initially responsive patients and the lack of standardized biomarkers to identify "BRCAness" pose serious threats to the clinical advance of PARP inhibitors; a feeling is also emerging that a BRCA-centered perspective might have missed the influence of additional, not negligible and DNA repair-independent PARP contributions onto therapy outcome. While regulatory approval for PARP1/2 inhibitors is still pending, novel therapeutic opportunities are sprouting from different branches of the PARP family, although they remain immature for clinical extrapolation. This review is an endeavor to provide a comprehensive appraisal of the multifaceted biology of PARPs and their evolving impact on cancer therapeutics.
Collapse
Affiliation(s)
- Barbara Lupo
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Torino, Italy; Laboratory of Molecular Pharmacology, Candiolo Cancer Institute, FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Torino, Italy; Laboratory of Molecular Pharmacology, Candiolo Cancer Institute, FPO IRCCS, 10060 Candiolo, Torino, Italy.
| |
Collapse
|
21
|
Schoumacher M, Hurov KE, Lehár J, Yan-Neale Y, Mishina Y, Sonkin D, Korn JM, Flemming D, Jones MD, Antonakos B, Cooke VG, Steiger J, Ledell J, Stump MD, Sellers WR, Danial NN, Shao W. Inhibiting Tankyrases sensitizes KRAS-mutant cancer cells to MEK inhibitors via FGFR2 feedback signaling. Cancer Res 2014; 74:3294-305. [PMID: 24747911 DOI: 10.1158/0008-5472.can-14-0138-t] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tankyrases (TNKS) play roles in Wnt signaling, telomere homeostasis, and mitosis, offering attractive targets for anticancer treatment. Using unbiased combination screening in a large panel of cancer cell lines, we have identified a strong synergy between TNKS and MEK inhibitors (MEKi) in KRAS-mutant cancer cells. Our study uncovers a novel function of TNKS in the relief of a feedback loop induced by MEK inhibition on FGFR2 signaling pathway. Moreover, dual inhibition of TNKS and MEK leads to more robust apoptosis and antitumor activity both in vitro and in vivo than effects observed by previously reported MEKi combinations. Altogether, our results show how a novel combination of TNKS and MEK inhibitors can be highly effective in targeting KRAS-mutant cancers by suppressing a newly discovered resistance mechanism.
Collapse
Affiliation(s)
- Marie Schoumacher
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kristen E Hurov
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joseph Lehár
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yan Yan-Neale
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yuji Mishina
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dmitriy Sonkin
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joshua M Korn
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daisy Flemming
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael D Jones
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Brandon Antonakos
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vesselina G Cooke
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Janine Steiger
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jebediah Ledell
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark D Stump
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William R Sellers
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nika N Danial
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wenlin Shao
- Authors' Affiliations: Oncology Department, Novartis Institutes for BioMedical Research; Zalicus Inc., Cambridge; and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
22
|
Shim G, Ricoul M, Hempel WM, Azzam EI, Sabatier L. Crosstalk between telomere maintenance and radiation effects: A key player in the process of radiation-induced carcinogenesis. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2014; 760:S1383-5742(14)00002-7. [PMID: 24486376 PMCID: PMC4119099 DOI: 10.1016/j.mrrev.2014.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 01/14/2014] [Accepted: 01/22/2014] [Indexed: 02/06/2023]
Abstract
It is well established that ionizing radiation induces chromosomal damage, both following direct radiation exposure and via non-targeted (bystander) effects, activating DNA damage repair pathways, of which the proteins are closely linked to telomeric proteins and telomere maintenance. Long-term propagation of this radiation-induced chromosomal damage during cell proliferation results in chromosomal instability. Many studies have shown the link between radiation exposure and radiation-induced changes in oxidative stress and DNA damage repair in both targeted and non-targeted cells. However, the effect of these factors on telomeres, long established as guardians of the genome, still remains to be clarified. In this review, we will focus on what is known about how telomeres are affected by exposure to low- and high-LET ionizing radiation and during proliferation, and will discuss how telomeres may be a key player in the process of radiation-induced carcinogenesis.
Collapse
|
23
|
Abstract
Telomeres use distinct mechanisms (not used by arms or centromeres) to mediate cohesion between sister chromatids. However, the motivation for a specialized mechanism at telomeres is not well understood. Here we show, using fluorescence in situ hybridization and live-cell imaging, that persistent sister chromatid cohesion at telomeres triggers a prolonged anaphase in normal human cells and cancer cells. Excess cohesion at telomeres can be induced by inhibition of tankyrase 1, a poly(ADP-ribose) polymerase that is required for resolution of telomere cohesion, or by overexpression of proteins required to establish telomere cohesion, the shelterin subunit TIN2 and the cohesin subunit SA1. Regardless of the method of induction, excess cohesion at telomeres in mitosis prevents a robust and efficient anaphase. SA1- or TIN2-induced excess cohesion and anaphase delay can be rescued by overexpression of tankyrase 1. Moreover, we show that primary fibroblasts, which accumulate excess telomere cohesion at mitosis naturally during replicative aging, undergo a similar delay in anaphase progression that can also be rescued by overexpression of tankyrase 1. Our study demonstrates that there are opposing forces that regulate telomere cohesion. The observation that cells respond to unresolved telomere cohesion by delaying (but not completely disrupting) anaphase progression suggests a mechanism for tolerating excess cohesion and maintaining telomere integrity. This attempt to deal with telomere damage may be ultimately futile for aging fibroblasts but useful for cancer cells.
Collapse
Affiliation(s)
- Mi Kyung Kim
- Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY 10016
| | | |
Collapse
|
24
|
Structural basis and selectivity of tankyrase inhibition by a Wnt signaling inhibitor WIKI4. PLoS One 2013; 8:e65404. [PMID: 23762361 PMCID: PMC3675114 DOI: 10.1371/journal.pone.0065404] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 04/25/2013] [Indexed: 11/19/2022] Open
Abstract
Recently a novel inhibitor of Wnt signaling was discovered. The compound, WIKI4, was found to act through tankyrase inhibition and regulate β-catenin levels in many cancer cell lines and human embryonic stem cells. Here we confirm that WIKI4 is a high potency tankyrase inhibitor and that it selectively inhibits tankyrases over other ARTD enzymes tested. The binding mode of the compound to tankyrase 2 was determined by protein X-ray crystallography to 2.4 Å resolution. The structure revealed a novel binding mode to the adenosine subsite of the donor NAD(+) binding groove of the catalytic domain. Our results form a structural basis for further development of potent and selective tankyrase inhibitors based on the WIKI4 scaffold.
Collapse
|
25
|
Oh S, Wang Y, Zimbric J, Hendrickson EA. Human LIGIV is synthetically lethal with the loss of Rad54B-dependent recombination and is required for certain chromosome fusion events induced by telomere dysfunction. Nucleic Acids Res 2012; 41:1734-49. [PMID: 23275564 PMCID: PMC3561972 DOI: 10.1093/nar/gks1326] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Classic non-homologous end joining (C-NHEJ) is the predominant DNA double-strand break repair pathway in humans. Although seven genes Ku70, Ku86, DNA-PKcs, Artemis, DNA Ligase IV (LIGIV), X-ray cross-complementing group 4 and XRCC4-like factor are required for C-NHEJ, several of them also have ancillary functions. For example, Ku70:Ku86 possesses an essential telomere maintenance activity. In contrast, LIGIV is believed to function exclusively in C-NHEJ. Moreover, a viable LIGIV-null human B-cell line and LIGIV-reduced patient cell lines have been described. Together, these observations suggest that LIGIV (and hence C-NHEJ), albeit important, is nonetheless dispensable, whereas Ku70:Ku86 and telomere maintenance are essential. To confirm this hypothesis, we inactivated LIGIV in the epithelial human cell line, HCT116. The resulting LIGIV-null cell line was viable, verifying that the gene and C-NHEJ are not essential. However, functional inactivation of RAD54B, a key homologous recombination factor, in the LIGIV-null background yielded no viable clones, suggesting that the combined absence of RAD54B/homologous recombination and C-NHEJ is synthetically lethal. Finally, we demonstrate that LIGIV is differentially required for certain chromosome fusion events induced by telomere dysfunction—used for those owing to the overexpression of a dominant negative version of telomere recognition factor 2, but not used for those owing to absence of Ku70:Ku86.
Collapse
Affiliation(s)
- Sehyun Oh
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
26
|
Riffell JL, Lord CJ, Ashworth A. Tankyrase-targeted therapeutics: expanding opportunities in the PARP family. Nat Rev Drug Discov 2012; 11:923-36. [PMID: 23197039 DOI: 10.1038/nrd3868] [Citation(s) in RCA: 217] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The poly(ADP-ribose) polymerase (PARP) protein superfamily has wide-ranging roles in cellular processes such as DNA repair and WNT signalling. Efforts to pharmacologically target PARP enzymes have largely focused on PARP1 and the closely related PARP2, but recent work highlighting the role of another family member, tankyrase 1 (TANK1; also known as PARP5A and ARTD5), in the control of WNT signalling has fuelled interest in the development of additional inhibitors to target this enzyme class. Tankyrase function is also implicated in other processes such as the regulation of telomere length, lung fibrogenesis and myelination, suggesting that tankyrase inhibitors could have broad clinical utility. Here, we discuss the biology of tankyrases and the discovery of tankyrase-specific inhibitors. We also consider the challenges that lie ahead for the clinical development of PARP family inhibitors in general.
Collapse
Affiliation(s)
- Jenna L Riffell
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | | | | |
Collapse
|
27
|
Kim MK, Dudognon C, Smith S. Tankyrase 1 regulates centrosome function by controlling CPAP stability. EMBO Rep 2012; 13:724-32. [PMID: 22699936 DOI: 10.1038/embor.2012.86] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 05/18/2012] [Accepted: 05/23/2012] [Indexed: 01/05/2023] Open
Abstract
CPAP--a gene mutated in primary microcephaly--is required for procentriole formation. Here we show that CPAP degradation and function is controlled by the poly(ADP-ribose) polymerase tankyrase 1. CPAP is PARsylated by tankyrase 1 in vitro and in vivo. Overexpression of tankyrase 1 leads to CPAP proteasomal degradation, preventing centriole duplication, whereas depletion of tankyrase 1 stabilizes CPAP in G1, generating elongated procentrioles and multipolarity. Tankyrase 1 localizes to centrosomes exclusively in G1, coinciding with CPAP degradation. Hence, tankyrase 1-mediated PARsylation regulates CPAP levels during the cell cycle to limit centriole elongation and ensure normal centrosome function.
Collapse
Affiliation(s)
- Mi Kyung Kim
- Molecular Pathogenesis Program and Department of Pathology, Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
28
|
Le Chalony C, Hoffschir F, Gauthier LR, Gross J, Biard DS, Boussin FD, Pennaneach V. Partial complementation of a DNA ligase I deficiency by DNA ligase III and its impact on cell survival and telomere stability in mammalian cells. Cell Mol Life Sci 2012; 69:2933-49. [PMID: 22460582 PMCID: PMC3417097 DOI: 10.1007/s00018-012-0975-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/12/2012] [Accepted: 03/14/2012] [Indexed: 01/08/2023]
Abstract
DNA ligase I (LigI) plays a central role in the joining of strand interruptions during replication and repair. In our current study, we provide evidence that DNA ligase III (LigIII) and XRCC1, which form a complex that functions in single-strand break repair, are required for the proliferation of mammalian LigI-depleted cells. We show from our data that in cells with either dysfunctional LigI activity or depleted of this enzyme, both LigIII and XRCC1 are retained on the chromatin and accumulate at replication foci. We also demonstrate that the LigI and LigIII proteins cooperate to inhibit sister chromatid exchanges but that only LigI prevents telomere sister fusions. Taken together, these results suggest that in cells with dysfunctional LigI, LigIII contributes to the ligation of replication intermediates but not to the prevention of telomeric instability.
Collapse
|
29
|
Mozdarani H. Biological complexities in radiation carcinogenesis and cancer radiotherapy: impact of new biological paradigms. Genes (Basel) 2012; 3:90-114. [PMID: 24704845 PMCID: PMC3899963 DOI: 10.3390/genes3010090] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 01/07/2012] [Accepted: 01/13/2012] [Indexed: 12/31/2022] Open
Abstract
Although radiation carcinogenesis has been shown both experimentally and epidemiologically, the use of ionizing radiation is also one of the major modalities in cancer treatment. Various known cellular and molecular events are involved in carcinogenesis. Apart from the known phenomena, there could be implications for carcinogenesis and cancer prevention due to other biological processes such as the bystander effect, the abscopal effect, intrinsic radiosensitivity and radioadaptation. Bystander effects have consequences for mutation initiated cancer paradigms of radiation carcinogenesis, which provide the mechanistic justification for low-dose risk estimates. The abscopal effect is potentially important for tumor control and is mediated through cytokines and/or the immune system (mainly cell-mediated immunity). It results from loss of growth and stimulatory and/or immunosuppressive factors from the tumor. Intrinsic radiosensitivity is a feature of some cancer prone chromosomal breakage syndromes such as ataxia telangectiasia. Radiosensitivity is manifested as higher chromosomal aberrations and DNA repair impairment is now known as a good biomarker for breast cancer screening and prediction of prognosis. However, it is not yet known whether this effect is good or bad for those receiving radiation or radiomimetic agents for treatment. Radiation hormesis is another major concern for carcinogenesis. This process which protects cells from higher doses of radiation or radio mimic chemicals, may lead to the escape of cells from mitotic death or apoptosis and put cells with a lower amount of damage into the process of cancer induction. Therefore, any of these biological phenomena could have impact on another process giving rise to genome instability of cells which are not in the field of radiation but still receiving a lower amount of radiation. For prevention of radiation induced carcinogenesis or risk assessment as well as for successful radiation therapy, all these phenomena should be taken into account.
Collapse
Affiliation(s)
- Hossein Mozdarani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran.
| |
Collapse
|
30
|
Lamb RS, Citarelli M, Teotia S. Functions of the poly(ADP-ribose) polymerase superfamily in plants. Cell Mol Life Sci 2012; 69:175-89. [PMID: 21861184 PMCID: PMC11114847 DOI: 10.1007/s00018-011-0793-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/02/2011] [Accepted: 08/04/2011] [Indexed: 01/09/2023]
Abstract
Poly(ADP-ribosyl)ation is the covalent attachment of ADP-ribose subunits from NAD(+) to target proteins and was first described in plants in the 1970s. This post-translational modification is mediated by poly(ADP-ribose) polymerases (PARPs) and removed by poly(ADP-ribose) glycohydrolases (PARGs). PARPs have important functions in many biological processes including DNA repair, epigenetic regulation and transcription. However, these roles are not always associated with enzymatic activity. The PARP superfamily has been well studied in animals, but remains under-investigated in plants. Although plants lack the variety of PARP superfamily members found in mammals, they do encode three different types of PARP superfamily proteins, including a group of PARP-like proteins, the SRO family, that are plant specific. In plants, members of the PARP family and/or poly(ADP-ribosyl)ation have been linked to DNA repair, mitosis, innate immunity and stress responses. In addition, members of the SRO family have been shown to be necessary for normal sporophytic development. In this review, we summarize the current state of plant research into poly(ADP-ribosyl)ation and the PARP superfamily in plants.
Collapse
Affiliation(s)
- Rebecca S Lamb
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, 43210, USA.
| | | | | |
Collapse
|
31
|
Rad51 and DNA-PKcs are involved in the generation of specific telomere aberrations induced by the quadruplex ligand 360A that impair mitotic cell progression and lead to cell death. Cell Mol Life Sci 2011; 69:629-40. [PMID: 21773671 PMCID: PMC3265728 DOI: 10.1007/s00018-011-0767-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 06/16/2011] [Accepted: 06/30/2011] [Indexed: 11/18/2022]
Abstract
Functional telomeres are protected from non-homologous end-joining (NHEJ) and homologous recombination (HR) DNA repair pathways. Replication is a critical period for telomeres because of the requirement for reconstitution of functional protected telomere conformations, a process that involves DNA repair proteins. Using knockdown of DNA-PKcs and Rad51 expression in three different cell lines, we demonstrate the respective involvement of NHEJ and HR in the formation of telomere aberrations induced by the G-quadruplex ligand 360A during or after replication. HR contributed to specific chromatid-type aberrations (telomere losses and doublets) affecting the lagging strand telomeres, whereas DNA-PKcs-dependent NHEJ was responsible for sister telomere fusions as a direct consequence of G-quadruplex formation and/or stabilization induced by 360A on parental telomere G strands. NHEJ and HR activation at telomeres altered mitotic progression in treated cells. In particular, NHEJ-mediated sister telomere fusions were associated with altered metaphase-anaphase transition and anaphase bridges and resulted in cell death during mitosis or early G1. Collectively, these data elucidate specific molecular and cellular mechanisms triggered by telomere targeting by the G-quadruplex ligand 360A, leading to cancer cell death.
Collapse
|
32
|
Dregalla RC, Zhou J, Idate RR, Battaglia CLR, Liber HL, Bailey SM. Regulatory roles of tankyrase 1 at telomeres and in DNA repair: suppression of T-SCE and stabilization of DNA-PKcs. Aging (Albany NY) 2011; 2:691-708. [PMID: 21037379 PMCID: PMC2993799 DOI: 10.18632/aging.100210] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Intrigued by the dynamics of the seemingly contradictory yet integrated cellular responses to the requisites of preserving telomere integrity while also efficiently repairing damaged DNA, we investigated roles of the telomere associated poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) tankyrase 1 in both telomere function and the DNA damage response following exposure to ionizing radiation. Tankyrase 1 siRNA knockdown in human cells significantly elevated recombination specifically within telomeres, a phenotype with the potential of accelerating cellular senescence. Additionally, depletion of tankyrase 1 resulted in concomitant and rapid reduction of the nonhomologous end-joining protein DNA-PKcs, while Ku86 and ATM protein levels remained unchanged; DNA-PKcs mRNA levels were also unaffected. We found that the requirement of tankyrase 1 for DNA-PKcs protein stability reflects the necessity of its PARP enzymatic activity. We also demonstrated that depletion of tankyrase 1 resulted in proteasome-mediated DNA-PKcs degradation, explaining the associated defective damage response observed; i.e., increased sensitivity to ionizing radiation-induced cell killing, mutagenesis, chromosome aberration and telomere fusion. We provide the first evidence for regulation of DNA-PKcs by tankyrase 1 PARP activity and taken together, identify roles of tankyrase 1 with implications not only for DNA repair and telomere biology, but also for cancer and aging.
Collapse
Affiliation(s)
- Ryan C Dregalla
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, 80523-1618, USA
| | | | | | | | | | | |
Collapse
|
33
|
Diotti R, Loayza D. Shelterin complex and associated factors at human telomeres. Nucleus 2011; 2:119-35. [PMID: 21738835 PMCID: PMC3127094 DOI: 10.4161/nucl.2.2.15135] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 02/09/2011] [Accepted: 02/11/2011] [Indexed: 12/17/2022] Open
Abstract
The processes regulating telomere function have major impacts on fundamental issues in human cancer biology. First, active telomere maintenance is almost always required for full oncogenic transformation of human cells, through cellular immortalization by endowment of an infinite replicative potential. Second, the attrition that telomeres undergo upon replication is responsible for the finite replicative life span of cells in culture, a process called senescence, which is of paramount importance for tumor suppression in vivo. The process of telomere-based senescence is intimately coupled to the induction of a DNA damage response emanating from telomeres, which can be elicited by both the ATM and ATR dependent pathways. At telomeres, the shelterin complex is constituted by a group of six proteins which assembles quantitatively along the telomere tract, and imparts both telomere maintenance and telomere protection. Shelterin is known to regulate the action of telomerase, and to prevent inappropriate DNA damage responses at chromosome ends, mostly through inhibition of ATM and ATR. The roles of shelterin have increasingly been associated with transient interactions with downstream factors that are not associated quantitatively or stably with telomeres. Here, some of the important known interactions between shelterin and these associated factors and their interplay to mediate telomere functions are reviewed.
Collapse
Affiliation(s)
- Raffaella Diotti
- Department of Biological Sciences, Hunter College, New York, NY, USA
| | | |
Collapse
|
34
|
Poly(ADP-ribose) polymerase 3 (PARP3), a newcomer in cellular response to DNA damage and mitotic progression. Proc Natl Acad Sci U S A 2011; 108:2783-8. [PMID: 21270334 DOI: 10.1073/pnas.1016574108] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ADP ribosyl transferase [poly(ADP-ribose) polymerase] ARTD3(PARP3) is a newly characterized member of the ARTD(PARP) family that catalyzes the reaction of ADP ribosylation, a key posttranslational modification of proteins involved in different signaling pathways from DNA damage to energy metabolism and organismal memory. This enzyme shares high structural similarities with the DNA repair enzymes PARP1 and PARP2 and accordingly has been found to catalyse poly(ADP ribose) synthesis. However, relatively little is known about its in vivo cellular properties. By combining biochemical studies with the generation and characterization of loss-of-function human and mouse models, we describe PARP3 as a newcomer in genome integrity and mitotic progression. We report a particular role of PARP3 in cellular response to double-strand breaks, most likely in concert with PARP1. We identify PARP3 as a critical player in the stabilization of the mitotic spindle and in telomere integrity notably by associating and regulating the mitotic components NuMA and tankyrase 1. Both functions open stimulating prospects for specifically targeting PARP3 in cancer therapy.
Collapse
|
35
|
Citarelli M, Teotia S, Lamb RS. Evolutionary history of the poly(ADP-ribose) polymerase gene family in eukaryotes. BMC Evol Biol 2010; 10:308. [PMID: 20942953 PMCID: PMC2964712 DOI: 10.1186/1471-2148-10-308] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 10/13/2010] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The poly(ADP-ribose) polymerase (PARP) superfamily was originally identified as enzymes that catalyze the attachment of ADP-ribose subunits to target proteins using NAD+ as a substrate. The family is characterized by the catalytic site, termed the PARP signature. While these proteins can be found in a range of eukaryotes, they have been best studied in mammals. In these organisms, PARPs have key functions in DNA repair, genome integrity and epigenetic regulation. More recently it has been found that proteins within the PARP superfamily have altered catalytic sites, and have mono(ADP-ribose) transferase (mART) activity or are enzymatically inactive. These findings suggest that the PARP signature has a broader range of functions that initially predicted. In this study, we investigate the evolutionary history of PARP genes across the eukaryotes. RESULTS We identified in silico 236 PARP proteins from 77 species across five of the six eukaryotic supergroups. We performed extensive phylogenetic analyses of the identified PARPs. They are found in all eukaryotic supergroups for which sequence is available, but some individual lineages within supergroups have independently lost these genes. The PARP superfamily can be subdivided into six clades. Two of these clades were likely found in the last common eukaryotic ancestor. In addition, we have identified PARPs in organisms in which they have not previously been described. CONCLUSIONS Three main conclusions can be drawn from our study. First, the broad distribution and pattern of representation of PARP genes indicates that the ancestor of all extant eukaryotes encoded proteins of this type. Second, the ancestral PARP proteins had different functions and activities. One of these proteins was similar to human PARP1 and likely functioned in DNA damage response. The second of the ancestral PARPs had already evolved differences in its catalytic domain that suggest that these proteins may not have possessed poly(ADP-ribosyl)ation activity. Third, the diversity of the PARP superfamily is larger than previously documented, suggesting as more eukaryotic genomes become available, this gene family will grow in both number and type.
Collapse
Affiliation(s)
- Matteo Citarelli
- Plant Cellular and Molecular Biology Department, Ohio State University, 500 Aronoff Laboratory, 318 W. 12th Ave., Columbus, OH 43210 USA
| | - Sachin Teotia
- Plant Cellular and Molecular Biology Department, Ohio State University, 500 Aronoff Laboratory, 318 W. 12th Ave., Columbus, OH 43210 USA
- Molcular, Cellular and Developmental Biology Program, Ohio State University, Columbus, OH 43210 USA
| | - Rebecca S Lamb
- Plant Cellular and Molecular Biology Department, Ohio State University, 500 Aronoff Laboratory, 318 W. 12th Ave., Columbus, OH 43210 USA
- Molcular, Cellular and Developmental Biology Program, Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
36
|
Bombarde O, Boby C, Gomez D, Frit P, Giraud-Panis MJ, Gilson E, Salles B, Calsou P. TRF2/RAP1 and DNA-PK mediate a double protection against joining at telomeric ends. EMBO J 2010; 29:1573-84. [PMID: 20407424 DOI: 10.1038/emboj.2010.49] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 03/04/2010] [Indexed: 11/09/2022] Open
Abstract
DNA-dependent protein kinase (DNA-PK) is a double-strand breaks repair complex, the subunits of which (KU and DNA-PKcs) are paradoxically present at mammalian telomeres. Telomere fusion has been reported in cells lacking these proteins, raising two questions: how is DNA-PK prevented from initiating classical ligase IV (LIG4)-dependent non-homologous end-joining (C-NHEJ) at telomeres and how is the backup end-joining (EJ) activity (B-NHEJ) that operates at telomeres under conditions of C-NHEJ deficiency controlled? To address these questions, we have investigated EJ using plasmid substrates bearing double-stranded telomeric tracks and human cell extracts with variable C-NHEJ or B-NHEJ activity. We found that (1) TRF2/RAP1 prevents C-NHEJ-mediated end fusion at the initial DNA-PK end binding and activation step and (2) DNA-PK counteracts a potent LIG4-independent EJ mechanism. Thus, telomeres are protected against EJ by a lock with two bolts. These results account for observations with mammalian models and underline the importance of alternative non-classical EJ pathways for telomere fusions in cells.
Collapse
Affiliation(s)
- Oriane Bombarde
- Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Recent findings have thrust poly(ADP-ribose) polymerases (PARPs) into the limelight as potential chemotherapeutic targets. To provide a framework for understanding these recent observations, we review what is known about the structures and functions of the family of PARP enzymes, and then outline a series of questions that should be addressed to guide the rational development of PARP inhibitors as anticancer agents.
Collapse
Affiliation(s)
- Michèle Rouleau
- Laval University Medical Research Center, Laval University, Québec, Canada
| | | | | | | | | |
Collapse
|
38
|
Canudas S, Smith S. Differential regulation of telomere and centromere cohesion by the Scc3 homologues SA1 and SA2, respectively, in human cells. J Cell Biol 2009; 187:165-73. [PMID: 19822671 PMCID: PMC2768842 DOI: 10.1083/jcb.200903096] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 09/10/2009] [Indexed: 11/22/2022] Open
Abstract
Replicated sister chromatids are held together until mitosis by cohesin, a conserved multisubunit complex comprised of Smc1, Smc3, Scc1, and Scc3, which in vertebrate cells exists as two closely related homologues (SA1 and SA2). Here, we show that cohesin(SA1) and cohesin(SA2) are differentially required for telomere and centromere cohesion, respectively. Cells deficient in SA1 are unable to establish or maintain cohesion between sister telomeres after DNA replication in S phase. The same phenotype is observed upon depletion of the telomeric protein TIN2. In contrast, in SA2-depleted cells telomere cohesion is normal, but centromere cohesion is prematurely lost. We demonstrate that loss of telomere cohesion has dramatic consequences on chromosome morphology and function. In the absence of sister telomere cohesion, cells are unable to repair chromatid breaks and suffer sister telomere loss. Our studies elucidate the functional distinction between the Scc3 homologues in human cells and further reveal an essential role for sister telomere cohesion in genomic integrity.
Collapse
Affiliation(s)
- Silvia Canudas
- Molecular Pathogenesis Program and Department of Pathology, Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
39
|
Papeo G, Forte B, Orsini P, Perrera C, Posteri H, Scolaro A, Montagnoli A. Poly(ADP-ribose) polymerase inhibition in cancer therapy: are we close to maturity? Expert Opin Ther Pat 2009; 19:1377-400. [DOI: 10.1517/13543770903215883] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Williams R. Tankyrase: A new breed of telomere security guard. J Biophys Biochem Cytol 2009. [PMCID: PMC2654132 DOI: 10.1083/jcb.1844iti4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|