• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • Tagged with
  • 8
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

HTLV-1 bZIP factor suppresses TDP1 expression through inhibition of NRF-1 in adult T-cell leukemia / HTLV-1 bZIP factorは成人T細胞白血病においてNRF-1を阻害しTDP1発現を抑制する

Takiuchi, Yoko 26 March 2018 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第20977号 / 医博第4323号 / 新制||医||1026(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 小柳 義夫, 教授 小川 誠司, 教授 朝長 啓造 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
2

Abacavir, an anti-HIV-1 drug, targets TDP1-deficient adult T cell leukemia / 抗HIV薬アバカビルは、TDP1が欠損している成人T細胞白血病を標的とする

Tada, Kohei 24 November 2015 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第19363号 / 医博第4040号 / 新制||医||1011(附属図書館) / 32377 / 新制||医||1011 / 京都大学大学院医学研究科医学専攻 / (主査)教授 小柳 義夫, 教授 河本 宏, 教授 松岡 雅雄 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
3

High Temperature Drives Topoisomerase Mediated Chromosomal Break Repair Pathway Choice.

Ashour, M.E., Allam, W., Elsayed, W., Atteya, R., Elserafy, M., Magdeldin, S., Hassan, M.K., El-Khamisy, Sherif 01 November 2023 (has links)
Yes / Cancer-causing mutations often arise from inappropriate DNA repair, yet acute exposure to DNA damage is widely used to treat cancer. The challenge remains in how to specifically induce excessive DNA damage in cancer cells while minimizing the undesirable effects of genomic instability in noncancerous cells. One approach is the acute exposure to hyperthermia, which suppresses DNA repair and synergizes with radiotherapy and chemotherapy. An exception, however, is the protective effect of hyperthermia on topoisomerase targeting therapeutics. The molecular explanation for this conundrum remains unclear. Here, we show that hyperthermia suppresses the level of topoisomerase mediated single- and double-strand breaks induced by exposure to topoisomerase poisons. We further uncover that, hyperthermia suppresses hallmarks of genomic instability induced by topoisomerase targeting therapeutics by inhibiting nuclease activities, thereby channeling repair to error-free pathways driven by tyrosyl-DNA phosphodiesterases. These findings provide an explanation for the protective effect of hyperthermia from topoisomerase-induced DNA damage and may help to explain the inverse relationship between cancer incidence and temperature. They also pave the way for the use of controlled heat as a therapeutic adjunct to topoisomerase targeting therapeutics.
4

DNA double-strand break formation and signalling in response to transcription-blocking topoisomerase I complexes / Formation et signalisation des cassures double-brin de l'ADN lors d'un blocage de la transcription

Cristini, Agnese 13 November 2015 (has links)
La topoisomérase I (Top1) élimine les surenroulements de l'ADN générés lors de la transcription en produisant transitoirement des complexes de clivage Top1-ADN (Top1cc). Ces Top1cc transitoires peuvent être stabilisés par les camptothécines, dont sont dérivés des agents anticancéreux, et par les fréquentes altérations de l'ADN. Bien que les Top1cc stabilisés soient des lésions qui bloquent efficacement la transcription, la compréhension des processus moléculaires qui résultent du blocage des complexes transcriptionnels par les Top1cc est encore limitée. Des travaux précédents ont montré que les Top1cc stabilisés produisent des cassures double-brin (DSBs) de l'ADN dépendantes de la transcription qui activent ATM. Dans ce projet, nous avons utilisé des cellules quiescentes traitées avec la camptothécine pour induire des Top1cc bloquant la transcription et nous avons étudié les mécanismes de la production et de la signalisation des DSBs. Nous montrons que les DSBs sont produites préférentiellement dans les régions sub-télomériques lors de la réparation des Top1cc bloquant la transcription par les cassures simple-brin de l'ADN générées après la protéolyse de la Top1 et avant l'action de Tdp1. L'analyse de la signalisation de ces DSBs révèle une nouvelle fonction de DNA-PK dans la promotion de l'ubiquitinylation conduisant (i) à l'activité complète d'ATM aux sites des DSBs en favorisant l'ubiquitination d'H2AX et H2A, et (ii) à l'augmentation de la réparation des Top1cc en favorisant la protéolyse de la Top1. Enfin, nous montrons que les DSBs co-transcriptionnelles induisent la mort des cellules quiescentes. L'ensemble de ces résultats apportent un nouvel aperçu des réponses cellulaires aux camptothécines, et suggèrent que les DSBs qui résultent des Top1cc bloquant la transcription puissent contribuer à la pathogénèse du syndrome neurodégénératif SCAN1, qui est causé par une déficience en Tdp1. / Topoisomerase I (Top1) removes DNA supercoiling generated during transcription by producing Top1-DNA cleavage complexes (Top1cc). These transient Top1cc can be stabilized by camptothecins, from which anticancer drugs are derived, and by common DNA alterations. Although stabilized Top1cc are potent transcription-blocking lesions, our understanding regarding the molecular processes resulting from the stalling of transcription complexes by Top1cc is currently limited. Previous work showed that stabilized Top1cc produce transcription-dependent DNA double-strand breaks (DSBs) that activate ATM signalling. In this project, we used camptothecin-treated quiescent cells to induce transcription-blocking Top1cc and study the mechanisms of DSB production and signalling. We show that DSBs form preferentially at subtelomeric regions during the repair of transcription-blocking Top1cc from DNA single-strand breaks generated after Top1 proteolysis and before Tdp1 action. Analysis of DSB signalling reveals a novel function of DNA-PK in promoting protein ubiquitination leading (i) to full ATM activity at DSB sites by promoting H2AX and H2A ubiquitination, and (ii) to enhancement of Top1cc repair by promoting Top1 proteolysis. Finally, we show that co-transcriptional DSBs kill quiescent cells. Together, these findings provide new insights into the cellular responses to camptothecins and further suggest that DSBs arising from transcription-blocking Top1cc may contribute to the pathogenesis of the neurodegenerative SCAN1 syndrome, which is caused by Tdp1 deficiency.
5

ROLE OF TYROSYL-DNA PHOSPHODIESTERASE (TDP 1) ON REPAIR OF 3′-PHOSPHOGLYCOLATE (3′- PG) TERMINATED DNA DOUBLE-STRAND BREAKS (DSBS) AND IN RESPONSE TO OXIDATIVE STRESS

Zhou, Tong 29 November 2012 (has links)
DNA DSBs are most toxic to cells because they can lead to genomic rearrangements and even cell death. Most DSBs induced by ionizing radiation or radiomimetic drugs such as calicheamicin and bleomycin, bear 3′-phosphate or 3′- PG moieties that must be removed to allow subsequent gap filling and ligation. DSBs can be repaired by two main pathways: the homologous recombination (HR) pathway and the non-homologous end-joining (NHEJ) pathway, NHEJ is the primary repair pathway in mammalian cells. While HR repairs single strand breaks (SSBs) or DSBs accurately by using an undamaged copy of the sequence mostly at late S phase and G2 phase, the NHEJ pathway repairs DSBs without the requirement for sequence homology in a processing that may be error-free or error- prone and is most active at G1 phase. TDP1 is a DNA repair enzyme in both pathways, It associates with DNA SSB repair proteins XRCC1 and DNA ligase III and plays a role in processing of topoisomerase I- mediated SSBs. Our early results suggested that TDP1 also can remove protruding 3’- PG and other 3’ blocks from DSBs ends in vitro. A homozygous H493R mutation in the active site of TDP1 causes spinocerebellar ataxia with axonal neuropathy (SCAN1), a rare autosomal recessive genetic disease with neurological symptoms including peripheral neuropathy. DNA damage and misrepair can be determined by measuring the incidence of chromosomal aberrations such as rings, breaks, dicentrics, acentric fragments, and translocations in metaphase cells, and micronuclei in interphase cells. To assess the possible role of TDP1 in DSB repair in intact cells, the radiosensitivity of SCAN1 cells was determined by using a dose-fractionation method of irradiation. The data indicated that, when exposed to fractionated radiation doses, the SCAN1 cells were more sensitive than normal cells. Moreover, following treatment of cells with calicheamicin, SCAN1 cells showed a significantly higher incidence of dicentric chromosomes, acentric fragments, and micronuclei compared to normal cells, indicating that calicheamicin-induced DSBs were repaired less accurately and less efficiently, or more slowly in SCAN1 cells than in normal cells. All these results are consistent with a role for TDP1 in repair of 3’-PG DSBs in vivo. Oxidative stress is thought to induce replicative senescence and DNA damage in mouse embryo fibroblasts (MEFs). To determine the possible roles of oxidative stress on Tdp1-deficient MEFs, Tdp1-knockout MEFs and normal MEFs were cultured in 20% oxygen (atmospheric) and 3% (physiological) oxygen. The data from growth assays indicated that normal MEFs showed replicative senescence in 20% oxygen but not in 3% oxygen. Tdp1-knockout MEFs showed very poor growth compared to Tdp1 normal MEFs in both oxygen conditions, clearly suggesting an influence of repair of Tdp1 on oxidative stress induced DNA-DSBs in MEFs. Taken together, our results indicated that TDP1 is capable of removing protruding 3’-PG from DSB ends in intact cells. Moreover, DSBs induced by oxidative stress were repaired more slowly or inefficiently in MEFs when Tdp1 is absent, resulting in cell cycle arrest and poor cell growth.
6

Nonhomologous end-joining: TDP1-mediated processing, ATM-mediated signaling

Hawkins, Amy 13 November 2009 (has links)
This thesis investigates two separate features of nonhomologous end-joining (NHEJ) DNA repair: end processing, and DNA repair kinase signaling. DNA end processing was investigated in a mouse model of hereditary spinocerebellar ataxia with axonal neuropathy (SCAN1), a congenital neurodegenerative disease. SCAN1 is caused by a homozygous H493R mutation in the active site of tyrosyl-DNA phosphodiesterase (TDP1). To address how the H493R mutation elicits the specific pathologies of SCAN1 and to further elucidate the role of TDP1 in processing DNA end modifications, we generated a Tdp1 knockout mouse and characterized their behavior and specific repair deficiencies in extracts of embryonic fibroblasts from these animals. While Tdp1(-/-) mice appear phenotypically normal, extracts from Tdp1(-/-) fibroblasts exhibited deficiencies in processing 3'-phosphotyrosyl single-strand breaks and 3'-phosphoglycolate (PG) double-strand breaks (DSBs). Supplementing Tdp1(-/-) extracts with H493R TDP1 partially restored processing of 3'-phosphotyrosyl single-strand breaks, but with evidence of persistent covalent adducts between TDP1 and DNA, consistent with a proposed intermediate-stabilization effect of the SCAN1 mutation. However, H493R TDP1 supplementation had no effect on PG termini on 3' overhangs of DSBs; these remained completely unprocessed. Altogether, these results suggest that for 3'-PG overhang lesions, the SCAN1 mutation confers loss of function, while for 3'-phosphotyrosyl lesions, the mutation uniquely stabilizes a reaction intermediate. Furthermore, there is evidence that TDP1 also localizes to mitochondria, and mitochondrial DNA damage should not be excluded from significantly contributing to SCAN1 pathology. The effect of ATM signaling on NHEJ was investigated via a novel vector that allows for inducing I-SceI-mediated DNA DSBs that can then be analyzed for NHEJ repair events by fluorescence- and PCR-based methods. Using highly specific DNA kinase inhibitors and the repair cassette, we showed that inhibiting ATM reduced NHEJ by 80% in a U87 glioma model. Analysis of the PCR products from the NHEJ repair vector by PsiI restriction cleavage allowed for assessment of the fidelity of the NHEJ repair: inhibiting ATM reduced high-fidelity NHEJ by 40%. Together, these results suggest that ATM is critical for NHEJ of I-SceI DSBs and for high-fidelity repair, possibly due to ATM's effects on chromatin architecture surrounding the DSB.
7

Functional analysis of the DNA repair enzyme tyrosyl-DNA phosphodiesterase 1 (TDP1) in Trypanosoma brucei brucei

Carloni, Roberta January 2014 (has links)
In order to evaluate the suitability of the DNA repair enzyme tyrosyl-DNA phosphodiesterase 1 (TDP1) as a potential drug target for an anti-parasite therapy, we are studying its role in the bloodstream form of Trypanosoma brucei brucei, the eukaryotic parasite that causes African Sleeping Sickness. Eukaryotic TDP1 removes covalently trapped topoisomerase IB and other adducts from the 3’ end of the DNA at DNA strand breaks. Covalent topoisomerase IB stalling is caused by endogenous DNA damage and by anti-cancer drugs such as camptothecin (CPT). A potential approach could be to use TDP1 inhibitors synergistically with CPT in a combined anti-parasite therapy. T. brucei TDP1 knock out cells are hypersensitive to CPT and accumulate in the late S phase of the cell cycle upon treatment with the drug. The CPT hypersensitivity of the TDP1-/- cells can be fully rescued through ectopic expression of wild type TDP1. The catalytic activity of TDP1 is required for complementation of the CPT sensitivity since overexpression of a catalytically inactive mutant form of TDP1 further sensitises TDP1-/- cells to CPT. In this context, expression of the mutant H358N, which shows reduced activity, also increases sensitivity of TDP1-/- cells to the drug. Surprisingly, expressing TDP1 carrying an analogous mutation to the one that causes SCAN1, a human neurodegenerative disease, does not sensitise TDP1-/- cells further. With this unique set of mutant TDP1 proteins in a TDP1-/- background we hope to answer questions concerning TDP1 function that have so far been elusive.
8

PROCESSING OF 3′-BLOCKED DNA DOUBLE-STRAND BREAKS BY TYROSYL-DNA PHOSPHODIESTERASE 1, ARTEMIS AND POLYNUCLEOTIDE KINASE/ PHOSPHATASE

Kawale, Ajinkya S 01 January 2018 (has links)
DNA double-strand breaks (DSBs) containing unligatable termini are potent cytotoxic lesions leading to growth arrest or cell death. The Artemis nuclease and tyrosyl-DNA phosphodiesterase (TDP1) are each capable of resolving protruding 3′-phosphoglycolate (PG) termini of DNA double-strand breaks (DSBs). Consequently, a knockout of Artemis and a knockout/knockdown of TDP1 rendered cells sensitive to the radiomimetic agent neocarzinostatin (NCS), which induces 3′-PG-terminated DSBs. Unexpectedly, however, a knockdown or knockout of TDP1 in Artemis-null cells did not confer any greater sensitivity than either deficiency alone, indicating a strict epistasis between TDP1 and Artemis. Moreover, a deficiency in Artemis, but not TDP1, resulted in a fraction of unrepaired DSBs, which were assessed as 53BP1 foci. Conversely, a deficiency in TDP1, but not Artemis, resulted in a dramatic increase in dicentric chromosomes following NCS treatment. An inhibitor of DNA-dependent protein kinase, a key regulator of the classical nonhomologous end joining (C-NHEJ) pathway sensitized cells to NCS but eliminated the sensitizing effects of both TDP1 and Artemis deficiencies. Moreover, Polynucleotide Kinase/ Phosphatase (PNKP) is known to process 3′-phosphates and 5′-hydroxyls during DSB repair. PNKP-deficiency sensitized both HCT116 and HeLa cells to 3′-phosphate ended DSBs formed upon radiation and radiomimetic drug treatment. The increased cytotoxicity in the absence of PNKP was synonymous with persistent, un-rejoined 3′-phosphate-ended DSBs. However, DNA-PK deficiency sensitized PNKP-/- cells to low doses of NCS suggesting that, in the absence of PNKP, alternative enzyme(s) can remove 3′-phosphates in a DNA-PK-dependent manner. These results suggest that TDP1 and Artemis perform different functions in the repair of terminally blocked DSBs by the C-NHEJ pathway, and that whereas an Artemis deficiency prevents end joining of some DSBs, a TDP1 deficiency tends to promote DSB mis-joining. In addition, loss of PNKP significantly sensitizes cells to 3′-phosphate-ended DSBs due to a defect in 3′-dephosphorylation.

Page generated in 0.0342 seconds