1
|
Sze S, Bhardwaj A, Fnu P, Azarm K, Mund R, Ring K, Smith S. TERRA R-loops connect and protect sister telomeres in mitosis. Cell Rep 2023; 42:113235. [PMID: 37843976 PMCID: PMC10873023 DOI: 10.1016/j.celrep.2023.113235] [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: 04/27/2023] [Revised: 08/08/2023] [Accepted: 09/22/2023] [Indexed: 10/18/2023] Open
Abstract
Resolution of cohesion between sister telomeres in human cells depends on TRF1-mediated recruitment of the polyADP-ribosyltransferase tankyrase to telomeres. In human aged cells, due to insufficient recruitment of TRF1/tankyrase to shortened telomeres, sisters remain cohered in mitosis. This persistent cohesion plays a protective role, but the mechanism by which sisters remain cohered is not well understood. Here we show that telomere repeat-containing RNA (TERRA) holds sister telomeres together through RNA-DNA hybrid (R-loop) structures. We show that a tankyrase-interacting partner, the RNA-binding protein C19orf43, is required for repression of TERRA R-loops. Persistent telomere cohesion in C19orf43-depleted cells is counteracted by RNaseH1, confirming that RNA-DNA hybrids hold sisters together. Consistent with a protective role for persistent telomere cohesion, depletion of C19orf43 in aged cells reduces DNA damage and delays replicative senescence. We propose that the inherent inability of shortened telomeres to recruit R-loop-repressing machinery permits a controlled onset of senescence.
Collapse
Affiliation(s)
- Samantha Sze
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Priyanka Fnu
- University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | - Rachel Mund
- New York Medical College, Valhalla, NY 10595, USA
| | - Katherine Ring
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Susan Smith
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
2
|
Udroiu I, Marinaccio J, Sgura A. Inhibition of p53 and ATRX increases telomeric recombination in primary fibroblasts. FEBS Open Bio 2023; 13:1683-1698. [PMID: 37499040 PMCID: PMC10476563 DOI: 10.1002/2211-5463.13680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 07/29/2023] Open
Abstract
Telomere length can be maintained either by the telomerase enzyme or by alternative lengthening of telomeres (ALT), which is based on telomeric recombination. However, both mechanisms are inactive in most human somatic cells. ATRX has been previously identified as an ALT repressor gene. Nonetheless, TP53 is also deficient in most ALT cell lines, and previous works showed that it is an inhibitor of homologous recombination (HR). Despite this, the role of p53 as an ALT repressor has not been previously examined. Therefore, we investigated the effects of p53 and ATRX inhibition on normal human fibroblasts (devoid of any mutation), in the presence or absence of X-ray-induced telomeric damage. Performing immunofluorescence with antibodies for RAD51, H2AX, and TRF1 (for studying HR-mediated DNA damage repair) and CO-FISH (for telomeric sister chromatid exchanges), we observed that HR is a normal mechanism for the repair of telomeric damage, present also in noncancer cells. Moreover, we discovered that telomeric HR, as for HR in general, is significantly inhibited by p53. Indeed, we observed that inhibition of p53 drastically increases telomeric sister chromatid exchanges. We also confirmed that ATRX inhibition increases telomeric recombination. In particular, we observed an increase in crossover products, but a much higher increase in noncrossover products.
Collapse
Affiliation(s)
- Ion Udroiu
- Dipartimento di Scienze, Università "Roma Tre", Italy
| | | | | |
Collapse
|
3
|
Wilson C, Murnane JP. High-throughput screen to identify compounds that prevent or target telomere loss in human cancer cells. NAR Cancer 2022; 4:zcac029. [PMID: 36196242 PMCID: PMC9527662 DOI: 10.1093/narcan/zcac029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/09/2022] [Accepted: 09/29/2022] [Indexed: 11/14/2022] Open
Abstract
Chromosome instability (CIN) is an early step in carcinogenesis that promotes tumor cell progression and resistance to therapy. Using plasmids integrated adjacent to telomeres, we have previously demonstrated that the sensitivity of subtelomeric regions to DNA double-strand breaks (DSBs) contributes to telomere loss and CIN in cancer. A high-throughput screen was created to identify compounds that affect telomere loss due to subtelomeric DSBs introduced by I-SceI endonuclease, as detected by cells expressing green fluorescent protein (GFP). A screen of a library of 1832 biologically-active compounds identified a variety of compounds that increase or decrease the number of GFP-positive cells following activation of I-SceI. A curated screen done in triplicate at various concentrations found that inhibition of classical nonhomologous end joining (C-NHEJ) increased DSB-induced telomere loss, demonstrating that C-NHEJ is functional in subtelomeric regions. Compounds that decreased DSB-induced telomere loss included inhibitors of mTOR, p38 and tankyrase, consistent with our earlier hypothesis that the sensitivity of subtelomeric regions to DSBs is a result of inappropriate resection during repair. Although this assay was also designed to identify compounds that selectively target cells experiencing telomere loss and/or chromosome instability, no compounds of this type were identified in the current screen.
Collapse
Affiliation(s)
- Chris Wilson
- Department of Pharmaceutical Chemistry, Small Molecule Discovery Center, University of California, San Francisco, CA 94143, USA
| | - John P Murnane
- To whom correspondence should be addressed. Tel: +1 415 680 4434;
| |
Collapse
|
4
|
Khatun B, Kamath V, Sathyanarayana MB, Pai A, Gupta R, Malviya R. Emerging Role of Wnt/Beta-Catenin Signalling Pathways in Cancer Progression and Role of Small Molecule Tankyrase Inhibitors in Combating Multistage Cancers. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394717666210628122306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present review, an attempt has been made to summarize the development of various
Tankyrase inhibitors focussing on Wnt/beta-Catenin pathways along with other cancer targets.
The last decade witnessed a plethora of research related to the role of various genetic and epigenetic
events that are responsible for the progression of multistage cancers. As a result, the discovery of
various signalling pathways responsible for the development of different types of cancers has resulted
in the development of molecularly targeted anticancer agents. Out of the many signalling pathways,
the Wnt/beta-Catenin pathways have attracted the attention of many research groups owing
to their involvement in cell proliferation, role in apoptosis induction, cellular differentiation and also
cell migration. The abnormal activation of this pathways has been documented in a variety of tumour
cells. Another crucial factor that makes this pathway attractive to the researches is its direct
involvement with poly ADP ribose polymerases. Tankyrases are poly ADP (Adenosine Diphosphate)
ribose polymerases that have the capacity to inhibit Wnt/beta-Catenin pathways and become
an attractive target for anticancer drugs.
Collapse
Affiliation(s)
- Babli Khatun
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka,India
| | - Venkatesh Kamath
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka,India
| | - Muddukrishna Badamane Sathyanarayana
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka,India
| | - Aravinda Pai
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka,India
| | - Ramji Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh,India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh,India
| |
Collapse
|
5
|
|
6
|
Azarm K, Bhardwaj A, Kim E, Smith S. Persistent telomere cohesion protects aged cells from premature senescence. Nat Commun 2020; 11:3321. [PMID: 32620872 PMCID: PMC7335080 DOI: 10.1038/s41467-020-17133-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 06/05/2020] [Indexed: 01/29/2023] Open
Abstract
Human telomeres are bound by the telomere repeat binding proteins TRF1 and TRF2. Telomere shortening in human cells leads to a DNA damage response that signals replicative senescence. While insufficient loading of TRF2 at shortened telomeres contributes to the DNA damage response in senescence, the contribution of TRF1 to senescence induction has not been determined. Here we show that counter to TRF2 deficiency-mediated induction of DNA damage, TRF1 deficiency serves a protective role to limit induction of DNA damage induced by subtelomere recombination. Shortened telomeres recruit insufficient TRF1 and as a consequence inadequate tankyrase 1 to resolve sister telomere cohesion. Our findings suggest that the persistent cohesion protects short telomeres from inappropriate recombination. Ultimately, in the final division, telomeres are no longer able to maintain cohesion and subtelomere copying ensues. Thus, the gradual loss of TRF1 and concomitant persistent cohesion that occurs with telomere shortening ensures a measured approach to replicative senescence.
Collapse
Affiliation(s)
- Kameron Azarm
- Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Amit Bhardwaj
- Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Eugenie Kim
- Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Pathology, New York University School of Medicine, New York, NY, 10016, 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, 10016, USA.
| |
Collapse
|
7
|
Gambini A, Stein P, Savy V, Grow EJ, Papas BN, Zhang Y, Kenan AC, Padilla-Banks E, Cairns BR, Williams CJ. Developmentally Programmed Tankyrase Activity Upregulates β-Catenin and Licenses Progression of Embryonic Genome Activation. Dev Cell 2020; 53:545-560.e7. [PMID: 32442396 DOI: 10.1016/j.devcel.2020.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/16/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Embryonic genome activation (EGA) is orchestrated by an intrinsic developmental program initiated during oocyte maturation with translation of stored maternal mRNAs. Here, we show that tankyrase, a poly(ADP-ribosyl) polymerase that regulates β-catenin levels, undergoes programmed translation during oocyte maturation and serves an essential role in mouse EGA. Newly translated TNKS triggers proteasomal degradation of axin, reducing targeted destruction of β-catenin and promoting β-catenin-mediated transcription of target genes, including Myc. MYC mediates ribosomal RNA transcription in 2-cell embryos, supporting global protein synthesis. Suppression of tankyrase activity using knockdown or chemical inhibition causes loss of nuclear β-catenin and global reductions in transcription and histone H3 acetylation. Chromatin and transcriptional profiling indicate that development arrests prior to the mid-2-cell stage, mediated in part by reductions in β-catenin and MYC. These findings indicate that post-transcriptional regulation of tankyrase serves as a ligand-independent developmental mechanism for post-translational β-catenin activation and is required to complete EGA.
Collapse
Affiliation(s)
- Andrés Gambini
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Paula Stein
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Virginia Savy
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Edward J Grow
- Department of Oncological Sciences, Huntsman Cancer Institute and Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Brian N Papas
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yingpei Zhang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anna C Kenan
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Elizabeth Padilla-Banks
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Bradley R Cairns
- Department of Oncological Sciences, Huntsman Cancer Institute and Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Resolution of human ribosomal DNA occurs in anaphase, dependent on tankyrase 1, condensin II, and topoisomerase IIα. Genes Dev 2019; 33:276-281. [PMID: 30804226 PMCID: PMC6411013 DOI: 10.1101/gad.321836.118] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/07/2019] [Indexed: 11/25/2022]
Abstract
In this paper by Daniloski et al., the authors investigated the timing and mechanism of the resolution of the ribosomal DNA (rDNA) repeats during chromosome segregation in human cells. They found that resolution of human rDNA occurs in anaphase after the bulk of the genome, dependent on tankyrase 1, condensin II, and topoisomerase II. Formation of individualized sister chromatids is essential for their accurate segregation. In budding yeast, while most of the genome segregates at the metaphase to anaphase transition, resolution of the ribosomal DNA (rDNA) repeats is delayed. The timing and mechanism in human cells is unknown. Here we show that resolution of human rDNA occurs in anaphase after the bulk of the genome, dependent on tankyrase 1, condensin II, and topoisomerase IIα. Defective resolution leads to rDNA bridges, rDNA damage, and aneuploidy of an rDNA-containing acrocentric chromosome. Thus, temporal regulation of rDNA segregation is conserved between yeast and man and is essential for genome integrity.
Collapse
|
11
|
ADP-ribosylation and intracellular traffic: an emerging role for PARP enzymes. Biochem Soc Trans 2019; 47:357-370. [DOI: 10.1042/bst20180416] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
AbstractADP-ribosylation is an ancient and reversible post-translational modification (PTM) of proteins, in which the ADP-ribose moiety is transferred from NAD+ to target proteins by members of poly-ADP-ribosyl polymerase (PARP) family. The 17 members of this family have been involved in a variety of cellular functions, where their regulatory roles are exerted through the modification of specific substrates, whose identification is crucial to fully define the contribution of this PTM. Evidence of the role of the PARPs is now available both in the context of physiological processes and of cell responses to stress or starvation. An emerging role of the PARPs is their control of intracellular transport, as it is the case for tankyrases/PARP5 and PARP12. Here, we discuss the evidence pointing at this novel aspect of PARPs-dependent cell regulation.
Collapse
|
12
|
Kim MK. Novel insight into the function of tankyrase. Oncol Lett 2018; 16:6895-6902. [PMID: 30546421 PMCID: PMC6256358 DOI: 10.3892/ol.2018.9551] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/31/2018] [Indexed: 01/14/2023] Open
Abstract
Tankyrases are multifunctional poly(ADP-ribose) polymerases that regulate a variety of cellular processes, including Wnt signaling, telomere maintenance and mitosis regulation. Tankyrases interact with target proteins and regulate their interactions and stability through poly(ADP-ribosyl) ation. In addition to their roles in telomere maintenance and regulation of mitosis, tankyrase proteins regulate tumor suppressors, including AXIN, phosphatase and tensin homolog and angiomotin. Therefore, tankyrases may be effective targets for cancer treatment. Tankyrase inhibitors could affect a variety of carcinogenic pathways that promote uncontrolled proliferation, including Wnt, AKT, yes-associated protein, telomere maintenance and mitosis regulation. Recently, novel aspects of the function and mechanism of tankyrases have been reported, and a number of tankyrase inhibitors have been identified. A combination of conventional chemotherapy agents with tankyrase inhibitors may have synergistic anticancer effects. Therefore, it is expected that more advanced and improved tankyrase inhibitors will be developed, enabling novel therapeutic strategies against cancer and other tankyrase-associated diseases. The present review discusses tankyrase function and the role of tankyrase inhibitors in the treatment of cancer.
Collapse
Affiliation(s)
- Mi Kyung Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
13
|
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
|
14
|
Whole proteome analysis of human tankyrase knockout cells reveals targets of tankyrase-mediated degradation. Nat Commun 2017; 8:2214. [PMID: 29263426 PMCID: PMC5738441 DOI: 10.1038/s41467-017-02363-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/21/2017] [Indexed: 02/01/2023] Open
Abstract
Tankyrase 1 and 2 are poly(ADP-ribose) polymerases that function in pathways critical to cancer cell growth. Tankyrase-mediated PARylation marks protein targets for proteasomal degradation. Here, we generate human knockout cell lines to examine cell function and interrogate the proteome. We show that either tankyrase 1 or 2 is sufficient to maintain telomere length, but both are required to resolve telomere cohesion and maintain mitotic spindle integrity. Quantitative analysis of the proteome of tankyrase double knockout cells using isobaric tandem mass tags reveals targets of degradation, including antagonists of the Wnt/β-catenin signaling pathway (NKD1, NKD2, and HectD1) and three (Notch 1, 2, and 3) of the four Notch receptors. We show that tankyrases are required for Notch2 to exit the plasma membrane and enter the nucleus to activate transcription. Considering that Notch signaling is commonly activated in cancer, tankyrase inhibitors may have therapeutic potential in targeting this pathway. Tankyrase 1 and 2 are poly(ADP-ribose) polymerases that mark proteins for degradation, but there is a current lack of knowledge about their distinct functions and substrates. Here, the authors elucidate the cellular roles and substrates of these polymerases using comparative functional and proteomics analyses of tankyrase knockout cell lines.
Collapse
|
15
|
Mitotic Dysfunction Associated with Aging Hallmarks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:153-188. [DOI: 10.1007/978-3-319-57127-0_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Jiang T, Xu G, Wang Q, Yang L, Zheng L, Zhao J, Zhang X. In vitro expansion impaired the stemness of early passage mesenchymal stem cells for treatment of cartilage defects. Cell Death Dis 2017; 8:e2851. [PMID: 28569773 PMCID: PMC5520885 DOI: 10.1038/cddis.2017.215] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 02/08/2023]
Abstract
In vitro cultured autologous mesenchymal stem cells (MSCs) within passage 5 have been approved for clinical application in stem cell-based treatment of cartilage defects. However, their chondrogenic potential has not yet been questioned or verified. In this study, the chondrogenic potential of bone marrow MSCs at passage 3 (P3 BMSCs) was investigated both in cartilage repair and in vitro, with freshly isolated bone marrow mononuclear cells (BMMNCs) as controls. The results showed that P3 BMSCs were inferior to BMMNCs not only in their chondrogenic differentiation ability but also as candidates for long-term repair of cartilage defects. Compared with BMMNCs, P3 BMSCs presented a decay in telomerase activity and a change in chromosomal morphology with potential anomalous karyotypes, indicating senescence. In addition, interindividual variability in P3 BMSCs is much higher than in BMMNCs, demonstrating genomic instability. Interestingly, remarkable downregulation in cell cycle, DNA replication and mismatch repair (MMR) pathways as well as in multiple genes associated with telomerase activity and chromosomal stability were found in P3 BMSCs. This result indicates that telomerase and chromosome anomalies might originate from expansion, leading to impaired stemness and pluripotency of stem cells. In vitro culture and expansion are not recommended for cell-based therapy, and fresh BMMNCs are the first choice.
Collapse
Affiliation(s)
- Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Guojie Xu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, China
| | - Lihui Yang
- School of Nursing, Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Guangxi Biological Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Guangxi Biological Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
17
|
Nathubhai A, Haikarainen T, Koivunen J, Murthy S, Koumanov F, Lloyd MD, Holman GD, Pihlajaniemi T, Tosh D, Lehtiö L, Threadgill MD. Highly Potent and Isoform Selective Dual Site Binding Tankyrase/Wnt Signaling Inhibitors That Increase Cellular Glucose Uptake and Have Antiproliferative Activity. J Med Chem 2017; 60:814-820. [PMID: 27983846 DOI: 10.1021/acs.jmedchem.6b01574] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Compounds 13 and 14 were evaluated against 11 PARP isoforms to reveal that both 13 and 14 were more potent and isoform selective toward inhibiting tankyrases (TNKSs) than the "standard" inhibitor 1 (XAV939)5, i.e., IC50 = 100 pM vs TNKS2 and IC50 = 6.5 μM vs PARP1 for 14. In cellular assays, 13 and 14 inhibited Wnt-signaling, enhanced insulin-stimulated glucose uptake, and inhibited the proliferation of DLD-1 colorectal adenocarcinoma cells to a greater extent than 1.
Collapse
Affiliation(s)
- Amit Nathubhai
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath, Somerset BA2 7AY, U. K
| | - Teemu Haikarainen
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Jarkko Koivunen
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Sudarshan Murthy
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Françoise Koumanov
- Department of Biology and Biochemistry, University of Bath , Bath BA2 7AY, U. K
| | - Matthew D Lloyd
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath, Somerset BA2 7AY, U. K
| | - Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath , Bath BA2 7AY, U. K
| | - Taina Pihlajaniemi
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - David Tosh
- Department of Biology and Biochemistry, University of Bath , Bath BA2 7AY, U. K
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Michael D Threadgill
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath, Somerset BA2 7AY, U. K
| |
Collapse
|
18
|
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
|
19
|
Investigating the Interplay between Sister Chromatid Cohesion and Homolog Pairing in Drosophila Nuclei. PLoS Genet 2016; 12:e1006169. [PMID: 27541002 PMCID: PMC4991795 DOI: 10.1371/journal.pgen.1006169] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/14/2016] [Indexed: 11/19/2022] Open
Abstract
Following DNA replication, sister chromatids must stay connected for the remainder of the cell cycle in order to ensure accurate segregation in the subsequent cell division. This important function involves an evolutionarily conserved protein complex known as cohesin; any loss of cohesin causes premature sister chromatid separation in mitosis. Here, we examined the role of cohesin in sister chromatid cohesion prior to mitosis, using fluorescence in situ hybridization (FISH) to assay the alignment of sister chromatids in interphase Drosophila cells. Surprisingly, we found that sister chromatid cohesion can be maintained in G2 with little to no cohesin. This capacity to maintain cohesion is widespread in Drosophila, unlike in other systems where a reduced dependence on cohesin for sister chromatid segregation has been observed only at specific chromosomal regions, such as the rDNA locus in budding yeast. Additionally, we show that condensin II antagonizes the alignment of sister chromatids in interphase, supporting a model wherein cohesin and condensin II oppose each other’s functions in the alignment of sister chromatids. Finally, because the maternal and paternal homologs are paired in the somatic cells of Drosophila, and because condensin II has been shown to antagonize this pairing, we consider the possibility that condensin II-regulated mechanisms for aligning homologous chromosomes may also contribute to sister chromatid cohesion. As cells grow, they replicate their DNA to give rise to two copies of each chromosome, known as sister chromatids, which separate from each other once the cell divides. To ensure that sister chromatids end up in different daughter cells, they are kept together from DNA replication until mitosis via a connection known as cohesion. A protein complex known as cohesin is essential for this process. Our work in Drosophila cells suggests that factors other than cohesin also contribute to sister chromatid cohesion in interphase. Additionally, we observed that the alignment of sister chromatids is regulated by condensin II, a protein complex involved in the compaction of chromosomes prior to division as well as the regulation of inter-chromosomal associations. These findings highlight that, in addition to their important individual functions, cohesin and condensin II proteins may interact to organize chromosomes over the course of the cell cycle. Finally, building on prior observations that condensin II is involved in the regulation of somatic homolog pairing in Drosophila, our work suggests that the mechanisms underlying homolog pairing may also contribute to sister chromatid cohesion.
Collapse
|
20
|
Benhamou Y, Picco V, Pagès G. The telomere proteins in tumorigenesis and clinical outcomes of oral squamous cell carcinoma. Oral Oncol 2016; 57:46-53. [PMID: 27208844 DOI: 10.1016/j.oraloncology.2016.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/29/2016] [Accepted: 04/12/2016] [Indexed: 12/16/2022]
Abstract
The "Hallmarks of Cancer" describe the ways by which cancer cells bypass homeostasis. Escape from replicative senescence is one of the earliest features of cancer cells. Maintenance of the telomeres through reactivation of telomerase was initially associated with replicative immortality in various cancers. The shelterin complex, a telomeric hexaprotein association, plays a key role in telomere maintenance and in the hallmarks of cancer. Some shelterin proteins are overexpressed in diverse cancers and can promote tumorigenesis in animal models. Shelterin can also have an impact on tumor size, tumor growth and resistance to treatment. Studies into the expression level of shelterin in oral squamous cell carcinoma (OSCC) report contradictory results. Moreover, the exact role of these proteins in OSCC tumorigenesis remains uncertain. In this review, we examined the data linking telomeres and hallmarks of OSCC. Furthermore, we examined the literature concerning telomeres and the clinical outcome of OSCC. Finally, we propose a model encompassing the role of shelterin proteins in oral tumorigenesis and treatment outcome.
Collapse
Affiliation(s)
- Y Benhamou
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice CNRS UMR 7284/INSERM U 1081, France; University of Nice Sophia Antipolis, Nice University Hospital, Odontology Department, Nice, France
| | - V Picco
- Centre Scientifique de Monaco, Biomedical Department, 8 Quai Antoine Ier, MC-98000 Monaco, Monaco
| | - G Pagès
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice CNRS UMR 7284/INSERM U 1081, France
| |
Collapse
|
21
|
Zhong L, Ding Y, Bandyopadhyay G, Waaler J, Börgeson E, Smith S, Zhang M, Phillips SA, Mahooti S, Mahata SK, Shao J, Krauss S, Chi NW. The PARsylation activity of tankyrase in adipose tissue modulates systemic glucose metabolism in mice. Diabetologia 2016; 59:582-91. [PMID: 26631215 DOI: 10.1007/s00125-015-3815-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/30/2015] [Indexed: 12/29/2022]
Abstract
AIMS/HYPOTHESIS Tankyrase (TNKS) is a ubiquitously expressed molecular scaffold that is implicated in diverse processes. The catalytic activity of TNKS modifies substrate proteins through poly-ADP-ribosylation (PARsylation) and is responsive to cellular energetic state. Global deficiency of the TNKS protein in mice accelerates glucose utilisation and raises plasma adiponectin levels. The aim of this study was to investigate whether the PARsylation activity of TNKS in adipocytes plays a role in systemic glucose homeostasis. METHODS To inhibit TNKS-mediated PARsylation, we fed mice with a diet containing the TNKS-specific inhibitor G007-LK. To genetically inactivate TNKS catalysis in adipocytes while preserving its function as a molecular scaffold, we used an adipocyte-selective Cre transgene to delete TNKS exons that encoded the catalytic domain at the C-terminus. Tissue-specific insulin sensitivity in mice was investigated using hyperinsulinaemic-euglycaemic clamps. To model adipose-liver crosstalk ex vivo, we applied adipocyte-conditioned media to hepatocytes and assessed the effect on gluconeogenesis. RESULTS The TNKS inhibitor G007-LK improved glucose tolerance and insulin sensitivity and promptly increased plasma adiponectin levels. In female mice, but not in male mice, adipocyte-selective genetic inactivation of TNKS catalysis improved hepatic insulin sensitivity and post-transcriptionally increased plasma adiponectin levels. Both pharmacological and genetic TNKS inhibition in female mouse-derived adipocytes induced a change in secreted factors to decrease gluconeogenesis in primary hepatocytes. CONCLUSIONS/INTERPRETATION Systemic glucose homeostasis is regulated by the PARsylation activity of TNKS in adipocytes. This regulation is mediated in part by adipocyte-secreted factors that modulate hepatic glucose production. Pharmacological TNKS inhibition could potentially be used to improve glucose tolerance.
Collapse
Affiliation(s)
- Linlin Zhong
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA
| | - Yun Ding
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA
| | - Gautam Bandyopadhyay
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA
| | - Jo Waaler
- Oslo University Hospital, Oslo, Norway
| | - Emma Börgeson
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA
| | - Susan Smith
- New York University School of Medicine, New York, NY, USA
| | - Mingchen Zhang
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA
- First Affiliated Hospital of Xinjiang Medical University, Xinjiang, People's Republic of China
| | - Susan A Phillips
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Sepi Mahooti
- Department of Pathology, University of California, San Diego, CA, USA
| | - Sushil K Mahata
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA
| | - Jianhua Shao
- Department of Pediatrics, University of California, San Diego, CA, USA
| | | | - Nai-Wen Chi
- VA San Diego Healthcare System, San Diego, CA, USA.
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093-0673, USA.
| |
Collapse
|
22
|
Haikarainen T, Waaler J, Ignatev A, Nkizinkiko Y, Venkannagari H, Obaji E, Krauss S, Lehtiö L. Development and structural analysis of adenosine site binding tankyrase inhibitors. Bioorg Med Chem Lett 2016; 26:328-333. [DOI: 10.1016/j.bmcl.2015.12.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 01/08/2023]
|
23
|
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
|
24
|
Exploration of the nicotinamide-binding site of the tankyrases, identifying 3-arylisoquinolin-1-ones as potent and selective inhibitors in vitro. Bioorg Med Chem 2015; 23:5891-908. [PMID: 26189030 DOI: 10.1016/j.bmc.2015.06.061] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/17/2022]
Abstract
Tankyrases-1 and -2 (TNKS-1 and TNKS-2) have three cellular roles which make them important targets in cancer. Using NAD(+) as a substrate, they poly(ADP-ribosyl)ate TRF1 (regulating lengths of telomeres), NuMA (facilitating mitosis) and axin (in wnt/β-catenin signalling). Using molecular modelling and the structure of the weak inhibitor 5-aminoiso quinolin-1-one, 3-aryl-5-substituted-isoquinolin-1-ones were designed as inhibitors to explore the structure-activity relationships (SARs) for binding and to define the shape of a hydrophobic cavity in the active site. 5-Amino-3-arylisoquinolinones were synthesised by Suzuki-Miyaura coupling of arylboronic acids to 3-bromo-1-methoxy-5-nitro-isoquinoline, reduction and O-demethylation. 3-Aryl-5-methylisoquinolin-1-ones, 3-aryl-5-fluoroisoquinolin-1-ones and 3-aryl-5-methoxyisoquinolin-1-ones were accessed by deprotonation of 3-substituted-N,N,2-trimethylbenzamides and quench with an appropriate benzonitrile. SAR around the isoquinolinone core showed that aryl was required at the 3-position, optimally with a para-substituent. Small meta-substituents were tolerated but groups in the ortho-positions reduced or abolished activity. This was not due to lack of coplanarity of the rings, as shown by the potency of 4,5-dimethyl-3-phenylisoquinolin-1-one. Methyl and methoxy were optimal at the 5-position. SAR was rationalised by modelling and by crystal structures of examples with TNKS-2. The 3-aryl unit was located in a large hydrophobic cavity and the para-substituents projected into a tunnel leading to the exterior. Potency against TNKS-1 paralleled potency against TNKS-2. Most inhibitors were highly selective for TNKSs over PARP-1 and PARP-2. A range of highly potent and selective inhibitors is now available for cellular studies.
Collapse
|
25
|
Haikarainen T, Krauss S, Lehtio L. Tankyrases: structure, function and therapeutic implications in cancer. Curr Pharm Des 2015; 20:6472-88. [PMID: 24975604 PMCID: PMC4262938 DOI: 10.2174/1381612820666140630101525] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/26/2014] [Indexed: 12/22/2022]
Abstract
Several cellular signaling pathways are regulated by ADP-ribosylation, a posttranslational modification catalyzed by members of the ARTD superfamily. Tankyrases are distinguishable from the rest of this family by their unique domain organization, notably the sterile alpha motif responsible for oligomerization and ankyrin repeats mediating protein-protein interactions. Tankyrases are involved in various cellular functions, such as telomere homeostasis, Wnt/β-catenin signaling, glucose metabolism, and cell cycle progression. In these processes, Tankyrases regulate the interactions and stability of target proteins by poly (ADP-ribosyl)ation. Modified proteins are subsequently recognized by the E3 ubiquitin ligase RNF146, poly-ubiquitinated and predominantly guided to 26S proteasomal degradation. Several small molecule inhibitors have been described for Tankyrases; they compete with the co-substrate NAD+ for binding to the ARTD catalytic domain. The recent, highly potent and selective inhibitors possess several properties of lead compounds and can be used for proof-of-concept studies in cancer and other Tankyrase linked diseases.
Collapse
Affiliation(s)
| | | | - Lari Lehtio
- SFI-CAST Biomedical Innovation Center, Unit for Cell Signaling, Oslo University Hospital, Forskningsparken, Gaustadalleen 21, 0349, Oslo, Norway.
| |
Collapse
|
26
|
Disruption of Wnt/β-Catenin Signaling and Telomeric Shortening Are Inextricable Consequences of Tankyrase Inhibition in Human Cells. Mol Cell Biol 2015; 35:2425-35. [PMID: 25939383 DOI: 10.1128/mcb.00392-15] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/28/2015] [Indexed: 01/05/2023] Open
Abstract
Maintenance of chromosomal ends (telomeres) directly contributes to cancer cell immortalization. The telomere protection enzymes belonging to the tankyrase (Tnks) subfamily of poly(ADP-ribose) polymerases (PARPs) have recently been shown to also control transcriptional response to secreted Wnt signaling molecules. Whereas Tnks inhibitors are currently being developed as therapeutic agents for targeting Wnt-related cancers and as modulators of Wnt signaling in tissue-engineering agendas, their impact on telomere length maintenance remains unclear. Here, we leveraged a collection of Wnt pathway inhibitors with previously unassigned mechanisms of action to identify novel pharmacophores supporting Tnks inhibition. A multifaceted experimental approach that included structural, biochemical, and cell biological analyses revealed two distinct chemotypes with selectivity for Tnks enzymes. Using these reagents, we revealed that Tnks inhibition rapidly induces DNA damage at telomeres and telomeric shortening upon long-term chemical exposure in cultured cells. On the other hand, inhibitors of the Wnt acyltransferase Porcupine (Porcn) elicited neither effect. Thus, Tnks inhibitors impact telomere length maintenance independently of their affects on Wnt/β-catenin signaling. We discuss the implications of these findings for anticancer and regenerative medicine agendas dependent upon chemical inhibitors of Wnt/β-catenin signaling.
Collapse
|
27
|
Hosler GA, Davoli T, Mender I, Litzner B, Choi J, Kapur P, Shay JW, Wang RC. A primary melanoma and its asynchronous metastasis highlight the role of BRAF, CDKN2A, and TERT. J Cutan Pathol 2015; 42:108-17. [PMID: 25407517 PMCID: PMC4470704 DOI: 10.1111/cup.12444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/21/2014] [Accepted: 11/12/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Alterations in pathways including BRAF, CDKN2A, and TERT contribute to the development of melanoma, but the sequence in which the genetic alterations occur and their prognostic significance remains unclear. To clarify the role of these pathways, we analyzed a primary melanoma and its metastasis. METHODS Immunohistochemistry for BRAF-V600E, Sanger sequencing of BRAF and the TERT promoter, fluorescence in-situ hybridization, and telomere analyses were performed on a primary melanoma and its asynchronous cerebellar metastasis. Using the log-rank test and Cox-proportional model, the cancer genome atlas (TCGA) cohort of melanomas was analyzed for the effect of BRAF mutation and CDKN2A loss on survival. RESULTS The primary melanoma expressed mutant BRAF-V600E and possessed a homozygous deletion of CDKN2A. In addition to these early defects, the metastatic lesion also possessed evidence of aneuploidy and an activating mutation of the TERT promoter. In the TCGA melanoma cohort, there was a non-significant trend toward poor prognosis in early stage cutaneous melanoma patients with concomitant BRAF mutation and CDKN2A loss. CONCLUSION BRAF mutation and CDKN2A loss occurred early and TERT promoter mutation later in a case of lethal metastatic melanoma. The effects of these pathways on survival warrant further investigation in early stage cutaneous melanoma patients.
Collapse
Affiliation(s)
- Gregory A. Hosler
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
- ProPath, Dallas, TX
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Teresa Davoli
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA
| | - Ilgen Mender
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Brandon Litzner
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
| | - Jaehyuk Choi
- Department of Dermatology, Yale Medical School, New Haven, CT
| | - Payal Kapur
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Jerry W. Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Richard C. Wang
- Department of Dermatology, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
28
|
Affiliation(s)
- Helder Maiato
- Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular, Universidade do Porto; Porto Portugal
- Cell Division Unit, Department of Experimental Biology; Faculdade de Medicina, Universidade do Porto; Porto Portugal
| | - Olga Afonso
- Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular, Universidade do Porto; Porto Portugal
- Cell Division Unit, Department of Experimental Biology; Faculdade de Medicina, Universidade do Porto; Porto Portugal
| | - Irina Matos
- Chromosome Instability & Dynamics Laboratory; Instituto de Biologia Molecular e Celular, Universidade do Porto; Porto Portugal
- Cell Division Unit, Department of Experimental Biology; Faculdade de Medicina, Universidade do Porto; Porto Portugal
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Aredia F, Scovassi AI. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death. Biochem Pharmacol 2014; 92:157-63. [PMID: 24976506 DOI: 10.1016/j.bcp.2014.06.021] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022]
Abstract
Poly(ADP-ribosylation) results from the conversion of NAD(+) into ADP-ribose and the following addition of ADP-ribose units to form polymers, further bound to acceptor proteins; once post-translationally ADP-ribosylated, proteins could change their function in basic processes. Poly(ADP-ribosylation) is activated under critical situations represented by DNA damage and cellular stress, and modulated in different paradigms of cell death. The hallmarks of the main death processes, i.e. apoptosis, parthanatos, necroptosis and autophagy, will be described, focusing on the role of poly(ADP-ribose) as a signaling molecule.
Collapse
Affiliation(s)
- Francesca Aredia
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Anna Ivana Scovassi
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, 27100 Pavia, Italy.
| |
Collapse
|
32
|
Lisaingo K, Uringa EJ, Lansdorp PM. Resolution of telomere associations by TRF1 cleavage in mouse embryonic stem cells. Mol Biol Cell 2014; 25:1958-68. [PMID: 24829382 PMCID: PMC4072570 DOI: 10.1091/mbc.e13-10-0564] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Telomere associations have been observed during key cellular processes such as mitosis, meiosis, and carcinogenesis and must be resolved before cell division to prevent genome instability. Here we establish that telomeric repeat-binding factor 1 (TRF1), a core component of the telomere protein complex, is a mediator of telomere associations in mammalian cells. Using live-cell imaging, we show that expression of TRF1 or yellow fluorescent protein (YFP)-TRF1 fusion protein above endogenous levels prevents proper telomere resolution during mitosis. TRF1 overexpression results in telomere anaphase bridges and aggregates containing TRF1 protein and telomeric DNA. Site-specific protein cleavage of YFP-TRF1 by tobacco etch virus protease resolves telomere aggregates, indicating that telomere associations are mediated by TRF1. This study provides novel insight into the formation and resolution of telomere associations.
Collapse
Affiliation(s)
- Kathleen Lisaingo
- Terry Fox Laboratory, BC Cancer Research Centre, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| | - Evert-Jan Uringa
- Terry Fox Laboratory, BC Cancer Research Centre, University of British Columbia, Vancouver, BC V5Z 1L3, CanadaEuropean Research Institute for the Biology of Ageing, University of Groningen, University Medical CentreGroningen, NL-9713 AV Groningen, Netherlands
| | - Peter M Lansdorp
- Terry Fox Laboratory, BC Cancer Research Centre, University of British Columbia, Vancouver, BC V5Z 1L3, CanadaEuropean Research Institute for the Biology of Ageing, University of Groningen, University Medical CentreGroningen, NL-9713 AV Groningen, Netherlands
| |
Collapse
|