48 results on '"Thomas Helleday"'
Search Results
2. STAT3 differential scanning fluorimetry and differential scanning light scattering assays: Addressing a missing link in the characterization of STAT3 inhibitor interactions
- Author
-
Iryna Kolosenko, Matthieu Desroses, Thomas Helleday, Sander Busker, Juan Astorga-Wells, Dan Grandér, Sanaz Attarha, Roman A. Zubarev, and Brent D. G. Page
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Light ,Clinical Biochemistry ,Pharmaceutical Science ,Context (language use) ,Peptide ,Fluorescence spectroscopy ,Analytical Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Drug Development ,Protein Domains ,Drug Discovery ,Scattering, Radiation ,Fluorometry ,Electrophoretic mobility shift assay ,Binding site ,STAT3 ,Spectroscopy ,chemistry.chemical_classification ,Binding Sites ,biology ,Protein Stability ,Chemistry ,Temperature ,Small molecule ,Recombinant Proteins ,Cyclic S-Oxides ,High-Throughput Screening Assays ,030104 developmental biology ,030220 oncology & carcinogenesis ,Biophysics ,biology.protein ,Peptides ,Fluorescence anisotropy ,Protein Binding - Abstract
STAT3 protein is an established target for the development of new cancer therapeutic agents. Despite lacking a traditional binding site for small molecule inhibitors, many STAT3 inhibitors have been identified and explored for their anti-cancer activity. Because STAT3 signaling is mediated by protein-protein interactions, indirect methods are often employed to determine if proposed STAT3 inhibitors bind to STAT3 protein. While established STAT3 inhibition assays (such as the fluorescence polarization assay, electrophoretic mobility shift assay and ELISAs) have been used to identify novel inhibitors of STAT3 signaling, methods that directly assess STAT3 protein-inhibitor interactions could facilitate the development of novel inhibitors. In this context, we herein report new STAT3 binding assays based on differential scanning fluorimetry (DSF) and differential scanning light scattering (DSLS) to characterize interactions between STAT3 protein and inhibitors. Several peptide and small molecule STAT3 inhibitors have been evaluated, and new insight into how these compounds may interact with STAT3 is provided.
- Published
- 2018
- Full Text
- View/download PDF
3. Diverse heterocyclic scaffolds as dCTP pyrophosphatase 1 inhibitors. Part 2: Pyridone- and pyrimidinone-derived systems
- Author
-
Kristmundur Sigmundsson, Lars Johansson, Ulf Martens, Thomas Helleday, Annika Jenmalm Jensen, Maria Häggblad, Thomas Lundbäck, Bo Lundgren, Olga Loseva, Sabin Llona-Minguez, Ann-Sofie Jemth, and Martin Scobie
- Subjects
0301 basic medicine ,Pyridones ,Stereochemistry ,Clinical Biochemistry ,Chemical biology ,Pharmaceutical Science ,Pyrimidinones ,Ligands ,Biochemistry ,2-Pyridone ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,Humans ,Pyrimidone ,DCTP pyrophosphatase 1 ,Enzyme Inhibitors ,Pyrophosphatases ,Molecular Biology ,Ligand efficiency ,biology ,Chemistry ,Organic Chemistry ,Nucleotide Metabolism ,030104 developmental biology ,biology.protein ,Molecular Medicine - Abstract
Two screening campaigns using commercial (Chembridge DiverSET) and proprietary (Chemical Biology Consortium Sweden, CBCS) compound libraries, revealed a number of pyridone- and pyrimidinone-derived systems as inhibitors of the human dCTP pyrophosphatase 1 (dCTPase). In this letter, we present their preliminary structure-activity-relationships (SAR) and ligand efficiency scores (LE and LLE).
- Published
- 2017
- Full Text
- View/download PDF
4. With me or against me: Tumor suppressor and drug resistance activities of SAMHD1
- Author
-
Ida Hed Myrberg, Juliane Kutzner, Cynthia B.J. Paulin, Nikolas Herold, Jan-Inge Henter, Torsten Schaller, Sean G. Rudd, Thale Kristin Olsen, Thomas Helleday, and Kumar Sanjiv
- Subjects
0301 basic medicine ,Antimetabolites, Antineoplastic ,Cancer Research ,Deoxyribonucleoside triphosphate ,Drug Resistance ,Decitabine ,Endogenous retrovirus ,Drug resistance ,Biology ,Guanosine triphosphate ,Nervous System Malformations ,SAM Domain and HD Domain-Containing Protein 1 ,03 medical and health sciences ,chemistry.chemical_compound ,Autoimmune Diseases of the Nervous System ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Proliferation ,Monomeric GTP-Binding Proteins ,Tumor Suppressor Proteins ,Cytarabine ,Cell Biology ,Hematology ,medicine.disease ,Survival Analysis ,Virology ,Leukemia ,Treatment Outcome ,030104 developmental biology ,chemistry ,Leukemia, Myeloid ,Acute Disease ,Mutation ,Azacitidine ,Cancer research ,Sterile alpha motif ,medicine.drug ,SAMHD1 - Abstract
Sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1) is a (deoxy)guanosine triphosphate (dGTP/GTP)-activated deoxyribonucleoside triphosphate (dNTP) triphosphohydrolase involved in cellular dNTP homoeostasis. Mutations in SAMHD1 have been associated with the hyperinflammatory disease Aicardi–Goutieres syndrome (AGS). SAMHD1 also limits cells' permissiveness to infection with diverse viruses, including human immunodeficiency virus (HIV-1), and controls endogenous retroviruses. Increasing evidence supports the role of SAMHD1 as a tumor suppressor. However, SAMHD1 also can act as a resistance factor to nucleoside-based chemotherapies by hydrolyzing their active triphosphate metabolites, thereby reducing response of various malignancies to these anticancer drugs. Hence, informed cancer therapies must take into account the ambiguous properties of SAMHD1 as both an inhibitor of uncontrolled proliferation and a resistance factor limiting the efficacy of anticancer treatments. Here, we provide evidence that SAMHD1 is a double-edged sword for patients with acute myelogenous leukemia (AML). Our time-dependent analyses of The Cancer Genome Atlas (TCGA) AML cohort indicate that high expression of SAMHD1, even though it critically limits the efficacy of high-dose ara-C therapy, might be associated with more favorable disease progression.
- Published
- 2017
- Full Text
- View/download PDF
5. Validation and development of MTH1 inhibitors for treatment of cancer
- Author
-
Therese Pham, Kumar Sanjiv, T. Meijer, Jonas Nilsson, Christina Kalderén, Rozbeh Jafari, Fabienne Z. Gaugaz, Torkild Visnes, Annika Lindqvist, Prasad B. Wakchaure, Aleksandr Manoilov, Per Artursson, Gianluca Maddalo, U. Warpman Berglund, Olov A. Wallner, Pawel Baranczewski, V. Nagpal, Azita Rasti, Aljona Saleh, Elisee Wiita, Sean G. Rudd, Ann-Sofie Jemth, Martin Henriksson, Ingrid Almlöf, Helge Gad, Camilla Göktürk, Tobias Koolmeister, Alexey Chernobrovkin, Evert Homan, Thomas Helleday, Vassilis G. Gorgoulis, Lars Bräutigam, Berglind Osk Einarsdottir, Brinton Seashore-Ludlow, Y. Kallberg, Olga Loseva, T-C Lee, Janne Lehtiö, Sanjay R. Borhade, Björn Platzack, Patrick Herr, Anne M. Filppula, Karl S. A. Vallin, Roman A. Zubarev, Ioannis S. Pateras, Martin Scobie, and Nicolaas G. M. Schipper
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,DNA damage ,Pharmacology ,Mice ,Pharmaceutical Sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,cancer ,Nucleotide ,Enzyme Inhibitors ,RNA, Small Interfering ,Pharmaceutical sciences ,reactive oxygen species ,chemistry.chemical_classification ,Cancer och onkologi ,Nucleotides ,business.industry ,NUDT1 Gene ,Deoxyguanosine ,Cancer ,DNA ,Hematology ,Farmaceutiska vetenskaper ,medicine.disease ,Xenograft Model Antitumor Assays ,small molecule inhibitors ,Small molecule ,Phosphoric Monoester Hydrolases ,MTH1 ,3. Good health ,DNA Repair Enzymes ,Pyrimidines ,030104 developmental biology ,Oncology ,chemistry ,8-Hydroxy-2'-Deoxyguanosine ,Cancer and Oncology ,Cancer cell ,business ,Oxidation-Reduction - Abstract
Background: Previously we showed cancer cells rely on the MTH1 protein to prevent incorporation of otherwise deadly oxidised nucleotides into DNA and we developed MTH1 inhibitors which selectively kill cancer cells. Recently several new and potent inhibitors of MTH1 were demonstrated to be non toxic to cancer cells challenging the utility of MTH1 inhibition as a target for cancer treatment. Material and methods: Human cancer cell lines were exposed in vitro to MTH1 inhibitors or depleted of MTH1 by siRNA or shRNA. 8 oxodG was measured by immunostaining and modified comet assay. Thermal Proteome profiling proteomics cellular thermal shift assays kinase and CEREP panel were used for target engagement mode of action and selectivity investigations of MTH1 inhibitors. Effect of MTH1 inhibition on tumour growth was explored in BRAF V600E mutated malignant melanoma patient derived xenograft and human colon cancer SW480 and HCT116 xenograft models. Results: Here we demonstrate that recently described MTH1 inhibitors which fail to kill cancer cells also fail to introduce the toxic oxidized nucleotides into DNA. We also describe a new MTH1 inhibitor TH1579 (Karonudib) an analogue of TH588 which is a potent selective MTH1 inhibitor with good oral availability and demonstrates excellent pharmacokinetic and anti cancer properties in vivo. Conclusion: We demonstrate that in order to kill cancer cells MTH1 inhibitors must also introduce oxidized nucleotides into DNA. Furthermore we describe TH1579 as a best in class MTH1 inhibitor which we expect to be useful in order to further validate the MTH1 inhibitor concept. Key words: MTH1 reactive oxygen species cancer small molecule inhibitors DNA damage
- Published
- 2016
- Full Text
- View/download PDF
6. Pathways controlling dNTP pools to maintain genome stability
- Author
-
Thomas Helleday, Sean G. Rudd, and Nicholas C.K. Valerie
- Subjects
0301 basic medicine ,Genome instability ,DNA Repair ,DNA damage ,DNA repair ,DNA polymerase ,Biochemistry ,Genomic Instability ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Humans ,Nucleotide ,Pyrophosphatases ,Molecular Biology ,chemistry.chemical_classification ,biology ,Genome, Human ,Nucleotides ,Nucleic Acid Precursors ,DNA ,Cell Biology ,Phosphoric Monoester Hydrolases ,Oxidative Stress ,DNA Repair Enzymes ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Human genome ,Oxidation-Reduction ,Nucleoside ,DNA Damage - Abstract
Artificially modified nucleotides, in the form of nucleoside analogues, are widely used in the treatment of cancers and various other diseases, and have become important tools in the laboratory to characterise DNA repair pathways. In contrast, the role of endogenously occurring nucleotide modifications in genome stability is little understood. This is despite the demonstration over three decades ago that the cellular DNA precursor pool is orders of magnitude more susceptible to modification than the DNA molecule itself. More recently, underscoring the importance of this topic, oxidation of the cellular nucleotide pool achieved through targeting the sanitation enzyme MTH1, appears to be a promising anti-cancer strategy. This article reviews our current understanding of modified DNA precursors in genome stability, with a particular focus upon oxidised nucleotides, and outlines some important outstanding questions.
- Published
- 2016
- Full Text
- View/download PDF
7. Timeless Interacts with PARP-1 to Promote Homologous Recombination Repair
- Author
-
Si, Xie, Oliver, Mortusewicz, Hoi Tang, Ma, Patrick, Herr, Randy Y C, Poon, Randy R Y, Poon, Thomas, Helleday, and Chengmin, Qian
- Subjects
Models, Molecular ,Timeless ,DNA damage ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,Cell Cycle Proteins ,Biology ,Crystallography, X-Ray ,Substrate Specificity ,chemistry.chemical_compound ,Humans ,DNA Breaks, Double-Stranded ,Binding site ,Homologous Recombination ,Molecular Biology ,Genetics ,Binding Sites ,DNA replication ,Intracellular Signaling Peptides and Proteins ,Recombinational DNA Repair ,Cell Biology ,Cell biology ,Establishment of sister chromatid cohesion ,chemistry ,Poly(ADP-ribose) Polymerases ,Protein Multimerization ,Homologous recombination ,DNA ,HeLa Cells - Abstract
Human Timeless helps stabilize replication forks during normal DNA replication and plays a critical role in activation of the S phase checkpoint and proper establishment of sister chromatid cohesion. However, it remains elusive whether Timeless is involved in the repair of damaged DNA. Here, we identify that Timeless physically interacts with PARP-1 independent of poly(ADP-ribosyl)ation. We present high-resolution crystal structures of Timeless PAB (PARP-1-binding domain) in free form and in complex with PARP-1 catalytic domain. Interestingly, Timeless PAB domain specifically recognizes PARP-1, but not PARP-2 or PARP-3. Timeless-PARP-1 interaction does not interfere with PARP-1 enzymatic activity. We demonstrate that rapid and transient accumulation of Timeless at laser-induced DNA damage sites requires PARP-1, but not poly(ADP-ribosyl)ation and that Timeless is co-trapped with PARP-1 at DNA lesions upon PARP inhibition. Furthermore, we show that Timeless and PARP-1 interaction is required for efficient homologous recombination repair.
- Published
- 2015
- Full Text
- View/download PDF
8. Addiction to MTH1 protein results in intense expression in human breast cancer tissue as measured by liquid chromatography-isotope-dilution tandem mass spectrometry
- Author
-
Leona D. Scanlan, Alessandro Tona, Pawel Jaruga, Mark S. Lowenthal, Miral Dizdaroglu, Thomas Helleday, Erdem Coskun, Ann-Sofie Jemth, and Olga Loseva
- Subjects
Programmed cell death ,Molecular Sequence Data ,Breast Neoplasms ,Biology ,Tandem mass spectrometry ,Bioinformatics ,Biochemistry ,chemistry.chemical_compound ,Downregulation and upregulation ,Tandem Mass Spectrometry ,Cell Line, Tumor ,medicine ,Humans ,Trypsin ,Amino Acid Sequence ,Molecular Biology ,Nitrogen Isotopes ,Hydrolysis ,DNA replication ,Cancer ,Cell Biology ,medicine.disease ,Phosphoric Monoester Hydrolases ,DNA Repair Enzymes ,chemistry ,Cancer cell ,Cancer research ,Female ,Cancer biomarkers ,Peptides ,DNA ,Chromatography, Liquid - Abstract
MTH1 protein sanitizes the nucleotide pool so that oxidized 2'-deoxynucleoside triphosphates (dNTPs) cannot be used in DNA replication. Cancer cells require MTH1 to avoid incorporation of oxidized dNTPs into DNA that results in mutations and cell death. Inhibition of MTH1 eradicates cancer, validating MTH1 as an anticancer target. By overexpressing MTH1, cancer cells may mediate cancer growth and resist therapy. To date, there is unreliable evidence suggesting that MTH1 is increased in cancer cells, and available methods to measure MTH1 levels are indirect and semi-quantitative. Accurate measurement of MTH1 in disease-free tissues and malignant tumors of patients may be essential for determining if the protein is truly upregulated in cancers, and for the development and use of MTH1 inhibitors in cancer therapy. Here, we present a novel approach involving liquid chromatography isotope-dilution tandem mass spectrometry to positively identify and accurately quantify MTH1 in human tissues. We produced full length N-15-labeled MTH1 and used it as an internal standard for the measurements. Following trypsin digestion, seven tryptic peptides of both MTH1 and N-15-MTH1 were identified by their full scan and product ion spectra. These peptides provided a statistically significant protein score that would unequivocally identify MTH1. Next, we identified and quantified MTH1 in human disease-free breast tissues and malignant breast tumors, and in four human cultured cell lines, three of which were cancer cells. Extreme expression of MTH1 in malignant breast tumors was observed, suggesting that cancer cells are addicted to MTH1 for their survival. The approach described is expected to be applicable to the measurement of MTH1 levels in malignant tumors vs. surrounding disease-free tissues in cancer patients. This attribute may help develop novel treatment strategies and MTH1 inhibitors as potential drugs, and guide therapies. Published by Elsevier B.V.
- Published
- 2015
- Full Text
- View/download PDF
9. Lysophosphatidic acid receptor (LPAR) modulators: The current pharmacological toolbox
- Author
-
Thomas Helleday, Artin Ghassemian, and Sabin Llona-Minguez
- Subjects
Drug discovery ,Motility ,Chemotaxis ,Cell Biology ,Biology ,Ligands ,Biochemistry ,Logical partition ,chemistry.chemical_compound ,Signalling ,chemistry ,Lysophosphatidic acid ,Animals ,Humans ,lipids (amino acids, peptides, and proteins) ,Lysophospholipids ,Receptors, Lysophosphatidic Acid ,biological phenomena, cell phenomena, and immunity ,Receptor ,Neuroscience ,Pharmacological Phenomena ,G protein-coupled receptor - Abstract
Lysophosphatidic acids (LPA) are key lipid-signalling molecules that regulate a remarkably diverse set of cellular events, such as motility, chemotaxis, cell cycle progression, viability, and wound healing. The physiological and pathophysiological consequences of LPA signalling are evident and misregulation of LPA signalling can lead to pathologies like cancer, atherosclerosis, ischaemia, and fibrosis. LPA exerts its biological actions mainly through several types of G protein-coupled receptors, some of which display opposing or redundant effects. For this reason, selective LPA receptor small-molecule ligands can shine light on LPA biology and present an exciting opportunity for drug discovery endeavours. This review provides insights into the detailed chemical nature and pharmacological profile of the small-molecules thus far developed as LPA receptor modulators, as well as information on the preparation of key pharmaceuticals. This summary will facilitate future research efforts and nurture collaboration between chemists and biologists working in this emerging field.
- Published
- 2015
- Full Text
- View/download PDF
10. Development and validation of method for TH588 and TH287, potent MTH1 inhibitors and new anti-cancer agents, for pharmacokinetic studies in mice plasma
- Author
-
Thomas Helleday, Camilla Göktürk, Ulrika Warpman-Berglund, Ingrid Granelli, and Aljona Saleh
- Subjects
Spectrometry, Mass, Electrospray Ionization ,Bioanalysis ,Electrospray ionization ,Clinical Biochemistry ,Cmax ,Pharmaceutical Science ,Antineoplastic Agents ,Pharmacology ,Mass spectrometry ,Gas Chromatography-Mass Spectrometry ,Analytical Chemistry ,Mice ,Pharmacokinetics ,Drug Discovery ,Animals ,Protein precipitation ,Spectroscopy ,Chemistry ,Selected reaction monitoring ,Reproducibility of Results ,Phosphoric Monoester Hydrolases ,Mice, Inbred C57BL ,Pyrimidines ,Female ,Monoisotopic mass ,Half-Life - Abstract
MTH1 is a protein that is required for cancer cell survival and is overexpressed in cancer cells. TH588 and TH287 are two new compounds that inhibit the MTH1 protein. The inhibitors were tested in pharmacokinetic studies on mice. A bioanalytical method was developed and validated for determination in mice plasma. The method was based on protein precipitation followed by LC-MS/MS analysis. The separation was performed on an Ascentis Express RP-Amide C18 column. The mass spectrometer was operated in positive electrospray ionization mode and the analytes were determined with multiple reaction monitoring (MRM). Abundant monoisotopic fragments were used for quantification. Two additional fragments were used for conformational analysis. The recovery of the compounds in plasma varied between 61 and 91% and the matrix effects were low and ranged between -3% and +2%. The method showed to be selective, linear, accurate and precise, and applicable for preclinical pharmacokinetic studies of TH588 and TH287 in mouse plasma. Half-life (T1/2) was ≤3.5h and maximum concentration (Cmax) ranged between 0.82 and 338μM for the different administration routes and compounds.
- Published
- 2015
- Full Text
- View/download PDF
11. PARP inhibitor receives FDA breakthrough therapy designation in castration resistant prostate cancer: beyond germline BRCA mutations
- Author
-
Thomas Helleday
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Breakthrough therapy ,Ubiquitin-Protein Ligases ,Poly(ADP-ribose) Polymerase Inhibitors ,Castration resistant ,Poly (ADP-Ribose) Polymerase Inhibitor ,Piperazines ,Germline ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Germline mutation ,Internal medicine ,medicine ,Humans ,Drug Approval ,Germ-Line Mutation ,BRCA2 Protein ,United States Food and Drug Administration ,business.industry ,Hematology ,medicine.disease ,United States ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,030220 oncology & carcinogenesis ,PARP inhibitor ,Phthalazines ,business - Published
- 2016
- Full Text
- View/download PDF
12. SETD2-Dependent Histone H3K36 Trimethylation Is Required for Homologous Recombination Repair and Genome Stability
- Author
-
Csanád Z. Bachrati, Nicholas B. La Thangue, Timothy C. Humphrey, Sophia X. Pfister, Gaëlle Legube, Thomas Helleday, François Aymard, Sovan Sarkar, Lykourgos-Panagiotis Zalmas, Sara Ahrabi, and Andrew C.G. Porter
- Subjects
DNA Repair ,DNA repair ,RAD51 ,Biology ,Transfection ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Genomic Instability ,Histones ,Histone H2A ,Histone methylation ,Histone code ,Humans ,Homologous Recombination ,Replication protein A ,lcsh:QH301-705.5 ,C130 Cell Biology ,Recombinational DNA Repair ,Histone-Lysine N-Methyltransferase ,DNA repair protein XRCC4 ,Molecular biology ,3. Good health ,lcsh:Biology (General) ,Histone methyltransferase ,C700 Molecular Biology, Biophysics and Biochemistry ,Rad51 Recombinase ,Protein Binding - Abstract
Summary Modulating chromatin through histone methylation orchestrates numerous cellular processes. SETD2-dependent trimethylation of histone H3K36 is associated with active transcription. Here, we define a role for H3K36 trimethylation in homologous recombination (HR) repair in human cells. We find that depleting SETD2 generates a mutation signature resembling RAD51 depletion at I-SceI-induced DNA double-strand break (DSB) sites, with significantly increased deletions arising through microhomology-mediated end-joining. We establish a presynaptic role for SETD2 methyltransferase in HR, where it facilitates the recruitment of C-terminal binding protein interacting protein (CtIP) and promotes DSB resection, allowing Replication Protein A (RPA) and RAD51 binding to DNA damage sites. Furthermore, reducing H3K36me3 levels by overexpressing KDM4A/JMJD2A, an oncogene and H3K36me3/2 demethylase, or an H3.3K36M transgene also reduces HR repair events. We propose that error-free HR repair within H3K36me3-decorated transcriptionally active genomic regions promotes cell homeostasis. Moreover, these findings provide insights as to why oncogenic mutations cluster within the H3K36me3 axis., Graphical Abstract, Highlights • A role for SETD2 in DSB resection and homologous recombination repair • Histone H3K36me3 is required for homologous recombination • SETD2 and RAD51 suppress mutations arising from microhomology-mediated end-joining • Mutations affecting H3K36me3 levels may promote tumorigenesis, The SETD2 gene encodes the histone H3K36 trimethyltransferase. Pfister et al. now show that human SETD2-dependent H3K36me3 maintains genome stability by promoting error-free DNA repair through homologous recombination (HR). Upon DNA damage, SETD2-depleted cells exhibit reduced DNA resection, impaired recruitment of early HR factors, and increased utilization of the error-prone microhomology-mediated end-joining repair pathway. Eliminating H3K36me3 by overexpressing the oncogene KDM4A also impairs HR. Thus, H3K36me3 suppresses tumorigenesis by promoting accurate DNA repair.
- Published
- 2014
- Full Text
- View/download PDF
13. Therapeutic implications of MTH1 inhibitor TH1579 in cutaneous malignant melanoma
- Author
-
Johan Hansson, Ishani Das, Veronica Höiom, Rainer Tuominen, Suzanne Egyhazi Brage, L. Pudelko, Thomas Helleday, Lars Bräutigam, U.W. Berglund, I. Almlöf, and Helge Gad
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Cell cycle checkpoint ,business.industry ,DNA repair ,Melanoma ,medicine.medical_treatment ,Cancer ,Hematology ,Immunotherapy ,medicine.disease ,Oncology ,medicine ,Cancer research ,Progression-free survival ,Skin cancer ,business - Abstract
Background Cutaneous malignant melanoma (CMM) is the main cause of skin cancer related deaths with approximately 230,000 new cases diagnosed worldwide annually. Advanced disease has poor prognosis due to unsuccessful treatment. BRAF inhibitors have shown promising clinical results with longer progression free survival but relapses are common and overall survival (OS) rates have been modest. Currently, there is a paucity of targeted treatment options for patients harboring NRAS mutation who have a worse OS compared to patients with wildtype NRAS. The DNA repair protein MTH1 sanitizes the dNTP pool by hydrolyzing harmful 8-oxodG which can be induced by reactive oxygen species (ROS). MTH1 is upregulated in several cancers including CMM. CMM has high levels of ROS and thus inhibition of MTH1 and other DNA repair proteins may be a plausible therapeutic strategy. Methods Here we used a panel of CMM cells, patient derived cells and an orthotopic zebrafish disease model to measure cell viability after treatment with MTH1 inhibitor TH1579 using real time imaging and luminescence. We performed AmpliSEQ analysis (patient tumor material), FACS, time lapse microscopy, immunofluorescence and western blot to do functional and molecular studies. Results We observed loss in cell viability and reduction in sphere growth upon treatment with TH1579 both in vitro and in vivo. FACS analysis confirmed induction of cell death in BRAF and NRAS mutant cell lines but not in the wild type cells, which showed induction of cell cycle arrest. Heterogeneous co-cultures of CMM cells were sensitive to TH1579, thus suggesting its broad clinical applicability. The BRAF mutant cells were further sensitized by co-treatment of TH1579 with BRAF inhibitor. We have also observed that CMM patients who are non-responders to immunotherapy have higher basal mRNA levels of MTH1 and other DNA repair genes. We are currently validating if this holds true at the protein levels as well. Conclusions We suggest that MTH1 has an impact on response to immunotherapy and inhibition of MTH1 might be an attractive strategy to partially overcome resistance to immunotherapy and as an alternate strategy for treatment of CMM patients independent of BRAF/NRAS mutational status. Legal entity responsible for the study Ulrika Warpman Berglund. Funding Swedish Cancer Society, Knut and Alice Wallenberg foundation, The Swedish Research Council, Torsten and Ragnar Soderberg Foundation, The Swedish Foundation for Strategic Research. Disclosure H. Gad: Shareholder / Stockholder / Stock options: Oxcia AB. L. Brautigam: Shareholder / Stockholder / Stock options: Oxcia AB. L. Pudelko: Shareholder / Stockholder / Stock options: Oxcia AB. I. Almlof: Shareholder / Stockholder / Stock options: Oxcia AB. T. Helleday: Shareholder / Stockholder / Stock options: Oxcia AB; Licensing / Royalties, Officer / Board of Directors: Thomas Helleday Foundation. U.W. Berglund: Shareholder / Stockholder / Stock options: Oxcia AB; Officer / Board of Directors: Thomas Helleday Foundation. All other authors have declared no conflicts of interest.
- Published
- 2019
- Full Text
- View/download PDF
14. A facile and efficient synthesis of tetrahydro-β-carbolines
- Author
-
Marie-Caroline Jacques-Cordonnier, Sabin Llona-Minguez, Armando Cázares-Körner, Martin Scobie, Sylvain A. Jacques, Thomas Helleday, Matthieu Desroses, and Tobias Koolmeister
- Subjects
chemistry.chemical_compound ,Chemistry ,Propane ,Organic Chemistry ,Drug Discovery ,Microwave irradiation ,Organic chemistry ,Biochemistry ,Acid anhydride ,Catalysis - Abstract
This Letter describes a convenient, efficient, and clean synthesis of various tetrahydro-β-carbolines catalyzed by propane phosphonic acid anhydride T3P® under microwave irradiation.
- Published
- 2013
- Full Text
- View/download PDF
15. Castration Therapy of Prostate Cancer Results in Downregulation of HIF-1α Levels
- Author
-
Torvald Granfors, Firas L.T. Al-Ubaidi, Thomas Helleday, Lars Egevad, and Niklas Schultz
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,medicine.medical_treatment ,Down-Regulation ,Radiation Tolerance ,Prostate cancer ,chemistry.chemical_compound ,Downregulation and upregulation ,Prostate ,Internal medicine ,Humans ,Medicine ,Radiology, Nuclear Medicine and imaging ,Orchiectomy ,Aged ,Aged, 80 and over ,Radiation ,business.industry ,Biopsy, Needle ,Prostatic Neoplasms ,Androgen Antagonists ,Middle Aged ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Androgen ,Cell Hypoxia ,Radiation therapy ,medicine.anatomical_structure ,Castration ,Hypoxia-inducible factors ,chemistry ,Leuprolide ,business ,Biomarkers - Abstract
Neoadjuvant androgen deprivation in combination with radiotherapy of prostate cancer is used to improve radioresponsiveness and local tumor control. Currently, the underlying mechanism is not well understood. Because hypoxia causes resistance to radiotherapy, we wanted to test whether castration affects the degree of hypoxia in prostate cancer.In 14 patients with locally advanced prostate cancer, six to 12 prostatic needle core biopsy specimens were taken prior to castration therapy. Bilateral orchidectomy was performed in 7 patients, and 7 were treated with a GnRH-agonist (leuprorelin). After castrationm two to four prostatic core biopsy specimens were taken, and the level of hypoxia-inducible factor-1α (HIF-1α) in cancer was determined by immunofluorescence.Among biopsy specimens taken before castration, strong HIF-1α expression (mean intensity above 30) was shown in 5 patients, weak expression (mean intensity 10-30) in 3 patients, and background levels of HIF-1α (mean intensity 0-10) in 6 patients. Downregulation of HIF-1α expression after castration was observed in all 5 patients with strong HIF-1α precastration expression. HIF-1α expression was also reduced in 2 of 3 patients with weak HIF-1α precastration expression.Our data suggest that neoadjuvant castration decreases tumor cell hypoxia in prostate cancer, which may explain increased radiosensitivity after castration.
- Published
- 2012
- Full Text
- View/download PDF
16. Chemotherapy-induced toxicity—a secondary effect caused by released DNA?
- Author
-
Thomas Helleday
- Subjects
business.industry ,DNA ,Hematology ,030204 cardiovascular system & hematology ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Oncology ,Chemotherapy induced ,chemistry ,030220 oncology & carcinogenesis ,Antineoplastic Combined Chemotherapy Protocols ,Toxicity ,Humans ,Medicine ,business - Published
- 2017
- Full Text
- View/download PDF
17. CHD1: a new treatment biomarker for recombination deficiency in castration resistant prostate cancer?
- Author
-
Thomas Helleday
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Castration resistant ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,Internal medicine ,medicine ,Humans ,Castration ,Recombination, Genetic ,business.industry ,Prostatic Neoplasms ,Original Articles ,Hematology ,medicine.disease ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,Biomarker (medicine) ,business ,Biomarkers - Published
- 2017
- Full Text
- View/download PDF
18. Transcription inhibition by 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) causes DNA damage and triggers homologous recombination repair in mammalian cells
- Author
-
Niklas Schultz, Ponnari Gottipati, Thomas Helleday, and Ivaylo Stoimenov
- Subjects
Hypoxanthine Phosphoribosyltransferase ,DNA Repair ,Transcription, Genetic ,DNA repair ,Health, Toxicology and Mutagenesis ,FLP-FRT recombination ,RAD51 ,CHO Cells ,Biology ,Genetic recombination ,Histones ,chemistry.chemical_compound ,Cricetulus ,Cricetinae ,Genetics ,Animals ,Enzyme Inhibitors ,Phosphorylation ,Molecular Biology ,Recombination, Genetic ,Microscopy, Confocal ,Molecular biology ,DNA-Binding Proteins ,Non-homologous end joining ,enzymes and coenzymes (carbohydrates) ,High-mobility group ,chemistry ,Mutation ,Rad51 Recombinase ,Homologous recombination ,Dichlororibofuranosylbenzimidazole ,5,6-Dichloro-1-beta-D-ribofuranosylbenzimidazole ,DNA Damage - Abstract
Transcription, replication and homologous recombination are intrinsically connected and it is well established that an increase of transcription is associated with an increase in homologous recombination. Here, we have studied how homologous recombination is affected during transcription inhibition by 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB), a compound that prevents activating phosphorylations of the RNA Pol II C-terminal domain. We identify that DRB triggers an increase in homologous recombination within the hprt gene as well as increasing RAD51 foci formation in mammalian cells. Furthermore, we find that DRB-induced transcriptional stress is associated with formation of the nuclear foci of the phosphorylated form of H2AX (γH2AX). We accounted that about 72% of RAD51 foci co-localized with the observed γH2AX foci. Interestingly, we find that XRCC3 mutated, homologous recombination defective cells are hypersensitive to the toxic effect of DRB and fail to form RAD51 foci. In conclusion, we show that DRB-induced transcription inhibition is associated with the formation of a lesion that triggers RAD51-dependent homologous recombination repair, required for survival under transcriptional stress.
- Published
- 2011
- Full Text
- View/download PDF
19. Repair pathways independent of the Fanconi anemia nuclear core complex play a predominant role in mitigating formaldehyde-induced DNA damage
- Author
-
Thomas Helleday, Natsuko Kondo, Ken Ohnishi, Małgorzata Z. Zdzienicka, Akihisa Takahashi, Larry H. Thompson, Noritomo Okamoto, Eiichiro Mori, Takeo Ohnishi, Yosuke Nakagawa, Taichi Noda, and Hideo Asada
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,DNA Repair ,DNA damage ,DNA repair ,DNA, Recombinant ,Biophysics ,CHO Cells ,Biology ,Biochemistry ,Histones ,Mice ,Cricetulus ,Fanconi anemia ,Cricetinae ,Formaldehyde ,hemic and lymphatic diseases ,FANCD2 ,medicine ,Animals ,Molecular Biology ,BRCA2 Protein ,Fanconi Anemia Complementation Group A Protein ,Fanconi Anemia Complementation Group D2 Protein ,Chinese hamster ovary cell ,Fanconi Anemia Complementation Group C Protein ,nutritional and metabolic diseases ,Cell Biology ,medicine.disease ,Molecular biology ,Fanconi Anemia Complementation Group Proteins ,FANCA ,Cell culture ,Homologous recombination ,DNA Damage - Abstract
The role of the Fanconi anemia (FA) repair pathway for DNA damage induced by formaldehyde was examined in the work described here. The following cell types were used: mouse embryonic fibroblast cell lines FANCA(-/-), FANCC(-/-), FANCA(-/-)C(-/-), FANCD2(-/-) and their parental cells, the Chinese hamster cell lines FANCD1 mutant (mt), FANCGmt, their revertant cells, and the corresponding wild-type (wt) cells. Cell survival rates were determined with colony formation assays after formaldehyde treatment. DNA double strand breaks (DSBs) were detected with an immunocytochemical γH2AX-staining assay. Although the sensitivity of FANCA(-/-), FANCC(-/-) and FANCA(-/-)C(-/-) cells to formaldehyde was comparable to that of proficient cells, FANCD1mt, FANCGmt and FANCD2(-/-) cells were more sensitive to formaldehyde than the corresponding proficient cells. It was found that homologous recombination (HR) repair was induced by formaldehyde. In addition, γH2AX foci in FANCD1mt cells persisted for longer times than in FANCD1wt cells. These findings suggest that formaldehyde-induced DSBs are repaired by HR through the FA repair pathway which is independent of the FA nuclear core complex.
- Published
- 2011
- Full Text
- View/download PDF
20. Human RECQL5 overcomes thymidine-induced replication stress
- Author
-
Thomas Helleday, Alastair S. H. Goldman, Katie N. Myers, Rachel Blundred, and Helen E. Bryant
- Subjects
DNA Replication ,Recombination, Genetic ,RecQ Helicases ,Blotting, Western ,Cell Cycle ,Ubiquitination ,DNA replication ,Fluorescent Antibody Technique ,Eukaryotic DNA replication ,Cell Biology ,Biology ,Pre-replication complex ,Biochemistry ,Molecular biology ,Cell biology ,DNA replication factor CDT1 ,Replication factor C ,Control of chromosome duplication ,Minichromosome maintenance ,biology.protein ,Humans ,Origin recognition complex ,Molecular Biology ,Thymidine - Abstract
Accurate DNA replication is essential to genome integrity and is controlled by five human RecQ helicases, of which at least three prevent cancer and ageing. Here, we have studied the role of RECQL5, which is the least characterised of the five human RecQ helicases. We demonstrate that overexpressed RECQL5 promotes survival during thymidine-induced slowing of replication forks in human cells. The RECQL5 protein relocates specifically to stalled replication forks and suppresses thymidine-induced RPA foci, CHK1 signalling, homologous recombination and gammaH2AX activation. It is unlikely that RECQL5 promotes survival through translesion synthesis as PCNA ubiquitylation is also reduced. Interestingly, we also found that overexpressing RECQL5 relieves cells of the cell cycle arrest normally imposed by thymidine, but without causing mutations. In conclusion, we propose that RECQL5 stabilises the replication fork allowing replication to overcome the effects of thymidine and complete the cell cycle.
- Published
- 2010
- Full Text
- View/download PDF
21. 1180 Effects of MTH1 inhibitor TH1579 on cutaneous melanoma
- Author
-
Ishani Das, Linda Pudelko, U.W. Berglund, Helge Gad, Thomas Helleday, Suzanne Egyhazi Brage, Rainer Tuominen, Johan Hansson, and Lars Bräutigam
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,business.industry ,Cutaneous melanoma ,Cancer research ,Medicine ,Cell Biology ,Dermatology ,business ,Molecular Biology ,Biochemistry - Published
- 2018
- Full Text
- View/download PDF
22. Hydroxyurea-Stalled Replication Forks Become Progressively Inactivated and Require Two Different RAD51-Mediated Pathways for Restart and Repair
- Author
-
Eva Petermann, Niklas Schultz, Natalia Issaeva, Manuel Luis Orta, Thomas Helleday, and Universidad de Sevilla. Departamento de Biología Celular
- Subjects
DNA re-replication ,DNA Replication ,DNA Repair ,Semiconservative replication ,Biology ,Pre-replication complex ,Article ,Replication fork protection ,S Phase ,Substrate Specificity ,Control of chromosome duplication ,Minichromosome maintenance ,Cell Line, Tumor ,Humans ,Hydroxyurea ,RNA, Small Interfering ,Replication protein A ,Molecular Biology ,DNA ,Cell Biology ,Virology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,Origin recognition complex ,Rad51 Recombinase ,DNA Damage - Abstract
Summary Faithful DNA replication is essential to all life. Hydroxyurea (HU) depletes the cells of dNTPs, which initially results in stalled replication forks that, after prolonged treatment, collapse into DSBs. Here, we report that stalled replication forks are efficiently restarted in a RAD51-dependent process that does not trigger homologous recombination (HR). The XRCC3 protein, which is required for RAD51 foci formation, is also required for replication restart of HU-stalled forks, suggesting that RAD51-mediated strand invasion supports fork restart. In contrast, replication forks collapsed by prolonged replication blocks do not restart, and global replication is rescued by new origin firing. We find that RAD51-dependent HR is triggered for repair of collapsed replication forks, without apparent restart. In conclusion, our data suggest that restart of stalled replication forks and HR repair of collapsed replication forks require two distinct RAD51-mediated pathways., Graphical Abstract Highlights ► RAD51 promotes replication fork restart after short hydroxyurea (HU) blocks ► RAD51-mediated fork restart does not create homologous recombination (HR) products ► Long HU blocks lead to fork collapse; replication is restarted by new origin firing ► Collapsed replication forks are repaired by classical RAD51-dependent HR
- Published
- 2010
- Full Text
- View/download PDF
23. Zinc Binding Catalytic Domain of Human Tankyrase 1
- Author
-
Lari Lehtiö, Martin Hammarström, Thomas Helleday, L.G. Dahlgren, Susanne van den Berg, Andreas Johansson, L. Holmberg-Schiavone, J. Weigelt, R. Collins, and Tobias Karlberg
- Subjects
Tankyrases ,Binding Sites ,Molecular Structure ,biology ,Poly ADP ribose polymerase ,Amino Acid Motifs ,Molecular Sequence Data ,Tankyrase-1 ,Zinc ,Telomere Homeostasis ,Biochemistry ,Structural Biology ,Catalytic Domain ,Drug Design ,Cancer cell ,biology.protein ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,Binding site ,Molecular Biology ,Mitosis ,Polymerase - Abstract
Tankyrases are recently discovered proteins implicated in many important functions in the cell including telomere homeostasis and mitosis. Tankyrase modulates the activity of target proteins through poly(ADP-ribosyl)ation, and here we report the structure of the catalytic poly(ADP-ribose) polymerase (PARP) domain of human tankyrase 1. This is the first structure of a PARP domain from the tankyrase subfamily. The present structure reveals that tankyrases contain a short zinc-binding motif, which has not been predicted. Tankyrase activity contributes to telomere elongation observed in various cancer cells and tankyrase inhibition has been suggested as a potential route for cancer therapy. In comparison with other PARPs, significant structural differences are observed in the regions lining the substrate-binding site of tankyrase 1. These findings will be of great value to facilitate structure-based design of selective PARP inhibitors, in general, and tankyrase inhibitors, in particular.
- Published
- 2008
- Full Text
- View/download PDF
24. RPA Mediates Recombination Repair During Replication Stress and Is Displaced from DNA by Checkpoint Signalling in Human Cells
- Author
-
Thomas Helleday, Kate Sleeth, Jiri Bartek, Jaroslaw Dziegielewski, Claus Storgaard Sørensen, and Natalia Issaeva
- Subjects
DNA Replication ,DNA Repair ,genetic processes ,RAD52 ,RAD51 ,Biology ,complex mixtures ,chemistry.chemical_compound ,Structural Biology ,Cell Line, Tumor ,Replication Protein A ,Humans ,Hydroxyurea ,CHEK1 ,Molecular Biology ,Replication protein A ,Nucleic Acid Synthesis Inhibitors ,Molecular biology ,Rad52 DNA Repair and Recombination Protein ,Chromatin ,Cell biology ,Enzyme Activation ,Genes, cdc ,enzymes and coenzymes (carbohydrates) ,chemistry ,Checkpoint Kinase 1 ,Rad51 Recombinase ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,Protein Kinases ,Ataxia telangiectasia and Rad3 related ,DNA ,DNA Damage ,Signal Transduction - Abstract
The replication protein A (RPA) is involved in most, if not all, nuclear metabolism involving single-stranded DNA. Here, we show that RPA is involved in genome maintenance at stalled replication forks by the homologous recombination repair system in humans. Depletion of the RPA protein inhibited the formation of RAD51 nuclear foci after hydroxyurea-induced replication stalling leading to persistent unrepaired DNA double-strand breaks (DSBs). We demonstrate a direct role of RPA in homology directed recombination repair. We find that RPA is dispensable for checkpoint kinase 1 (Chk1) activation and that RPA directly binds RAD52 upon replication stress, suggesting a direct role in recombination repair. In addition we show that inhibition of Chk1 with UCN-01 decreases dissociation of RPA from the chromatin and inhibits association of RAD51 and RAD52 with DNA. Altogether, our data suggest a direct role of RPA in homologous recombination in assembly of the RAD51 and RAD52 proteins. Furthermore, our data suggest that replacement of RPA with the RAD51 and RAD52 proteins is affected by checkpoint signalling.
- Published
- 2007
- Full Text
- View/download PDF
25. Homologous recombination is involved in repair of chromium-induced DNA damage in mammalian cells
- Author
-
Thomas Helleday, Songmin Ying, and Helen E. Bryant
- Subjects
Chromium ,DNA Repair ,Genotype ,DNA damage ,DNA repair ,Health, Toxicology and Mutagenesis ,Biology ,Models, Biological ,Cell Line ,Homology directed repair ,XRCC1 ,Cricetinae ,Genetics ,Animals ,Molecular Biology ,Replication protein A ,Recombination, Genetic ,Chromosome Breakage ,DNA repair protein XRCC4 ,Endonucleases ,Molecular biology ,DNA-Binding Proteins ,X-ray Repair Cross Complementing Protein 1 ,DNA mismatch repair ,Rad51 Recombinase ,DNA Damage ,Nucleotide excision repair - Abstract
Chromium is a potent human carcinogen, probably because of its well-documented genotoxic effects. Chromate (Cr[VI]) causes a wide range of DNA lesions, including DNA crosslinks and strand breaks, presumably due to the direct and indirect effects of DNA oxidation. Homologous recombination repair (HRR) is important for error-free repair of lesions occurring at replication forks. Here, we show that HR deficient cell lines irs1SF and V-C8, deficient in XRCC3 and BRCA2, respectively, are hypersensitive to Cr[VI], implicating this repair pathway in repair of Cr[VI] damage. Furthermore, we find that Cr[VI] causes DNA double-strand breaks and triggers both Rad51 foci formation and induction of HRR. Collectively, these data suggest that HRR is important in repair of Cr[VI]-induced DNA damage. In addition, we find that ERCC1, XRCC1 and DNA-PKcs defective cells are hypersensitive to Cr[VI], indicating that several repair pathways cooperate in repairing Cr[VI]-induced DNA damage.
- Published
- 2006
- Full Text
- View/download PDF
26. Screening for genotoxicity using the DRAG assay: investigation of halogenated environmental contaminants
- Author
-
Thomas Helleday, Annika Allkvist, Klaus Erixon, Fredrik Johansson, Robert Nilsson, Dag Jenssen, Anna Malmvärn, and Åke Bergman
- Subjects
Male ,inorganic chemicals ,Alkylating Agents ,animal structures ,DNA Repair ,DNA repair ,Mitomycin ,Health, Toxicology and Mutagenesis ,7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide ,CHO Cells ,medicine.disease_cause ,Cell Line ,Toxicology ,DNA Adducts ,Cricetinae ,Genetics ,medicine ,Animals ,Humans ,Molecular Structure ,Mutagenicity Tests ,Chemistry ,technology, industry, and agriculture ,Hydrogen Peroxide ,Contamination ,Oxidants ,Drag ,Ethyl Methanesulfonate ,Environmental chemistry ,biological sciences ,Carcinogens ,Biological Assay ,Environmental Pollutants ,human activities ,Genotoxicity ,Mutagens - Abstract
The DRAG test is a rapid high-throughput screening assay for detection of repairable adducts by growth inhibition of Chinese hamster ovary cells (CHO) characterized by different defects in DNA repair. A more pronounced growth inhibition caused by a certain DNA-reactive substance in a repair-deficient cell line (EM9, UV4 and UV5) as compared to wild-type cells (AA8) is interpreted as a consequence of their inability to repair induced DNA lesions. Thus, the use of such cell lines in the DRAG test may provide information of the type of DNA lesions induced by a certain genotoxic substance. To select optimal assay conditions, as well as to provide a mechanistic basis for interpreting the results, the model compounds benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), ethyl methanesulfonate (EMS), mitomycin C (MMC) and hydrogen peroxide (H2O2) were used. These agents can induce bulky adducts, alkyl adducts, cross-links and oxidative damage, respectively. The specificity of the DRAG test constitutes an important prerequisite for its practical use in a broader context. To assess this aspect, we have investigated the genotoxic and cytotoxic properties of a selection of metabolites of and isomers from polychlorinated biphenyls (PCB) and polybrominated diphenyl ethers (PBDE), along with a few other halogenated compounds. All these compounds have been detected as pollutants in the external environment, and for most of them there is no convincing evidence of mutagenicity from conventional assays. As could be predicted from their mode of action, BPDE, MMC, and EMS were all found to be more toxic in the repair-deficient cell lines compared with wild-type cells. The results with H2O2 were inconclusive, and the PCB metabolite 4,4'-diOH-CB80 only exhibited borderline activity, while all other halogenated compounds, or their metabolites, were found to be inactive. In conclusion, the DRAG assay could provide a robust and useful tool when screening large numbers of potentially genotoxic agents, while in addition providing mechanistic information. However, the usefulness of the selected cell lines to detect oxidative damage may be limited.
- Published
- 2004
- Full Text
- View/download PDF
27. DNA repair rate and etoposide (VP16) resistance of tumor cell subpopulations derived from a single human small cell lung cancer
- Author
-
Hans Skovgaard Poulsen, Thomas Helleday, Lone Petersen, Lasse Tengbjerg Hansen, Claus Storgaard Sørensen, Mogens Spang-Thomsen, and Cecilia Lundin
- Subjects
Pulmonary and Respiratory Medicine ,Cancer Research ,ATP Binding Cassette Transporter, Subfamily B ,Lung Neoplasms ,Saccharomyces cerevisiae Proteins ,DNA Repair ,DNA repair ,genetic processes ,RAD51 ,DNA-Activated Protein Kinase ,Protein Serine-Threonine Kinases ,Radioresistance ,Tumor Cells, Cultured ,medicine ,Humans ,Neoplasm ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Carcinoma, Small Cell ,Nuclear protein ,Etoposide ,DNA-PKcs ,Endodeoxyribonucleases ,biology ,Topoisomerase ,Nuclear Proteins ,DNA, Neoplasm ,medicine.disease ,Antineoplastic Agents, Phytogenic ,DNA-Binding Proteins ,DNA Topoisomerases, Type II ,Exodeoxyribonucleases ,Oncology ,Biochemistry ,Drug Resistance, Neoplasm ,health occupations ,Cancer research ,biology.protein ,Rad51 Recombinase ,Tumor Suppressor Protein p53 ,DNA Damage ,medicine.drug - Abstract
Two human small cell lung cancer (SCLC) subpopulations, CPH 54A, and CPH 54B, established from the same patient tumor by in vitro cloning, were investigated. The tumor was classified as intermediate-type SCLC. The cellular sensitivity to ionizing radiation (IR) was previously determined in the two sublines both in vivo and in vitro. Here we measured the etoposide (VP16) sensitivity together with the induction and repair of VP16- and IR-induced DNA double-strand breaks (DSBs). The two subpopulations were found to differ significantly in sensitivity to VP16, with the radioresistant 54B subline also being VP16 resistant. In order to explain the VP16 resistant phenotype several mechanisms where considered. The p53 status, P-glycoprotein, MRP, topoisomerase IIalpha, and Mre11 protein levels, as well as growth kinetics, provided no explanations of the observed VP16 resistance. In contrast, a significant difference in repair of both VP16- and IR-induced DSBs, together with a difference in the levels of the DSB repair proteins DNA-dependent protein kinase (DNA-PK(cs)) and RAD51 was observed. The VP16- and radioresistant 54B subline exhibited a pronounced higher repair rate of DSBs and higher protein levels of both DNA-PK(cs) and RAD51 compared with the sensitive 54A subline. We suggest, that different DSB repair rates among tumor cell subpopulations of individual SCLC tumors may be a major determinant for the variation in clinical treatment effect observed in human SCLC tumors of identical histological subtype.
- Published
- 2003
- Full Text
- View/download PDF
28. DNA double-strand breaks associated with replication forks are predominantly repaired by homologous recombination involving an exchange mechanism in mammalian cells11Edited by J. Karn
- Author
-
Cecilia Lundin, Thomas Helleday, and Catherine Arnaudeau
- Subjects
DNA repair ,DNA damage ,cells ,genetic processes ,fungi ,Mutant ,DNA replication ,Biology ,Molecular biology ,DNA-binding protein ,enzymes and coenzymes (carbohydrates) ,chemistry.chemical_compound ,chemistry ,Structural Biology ,Gene duplication ,biological phenomena, cell phenomena, and immunity ,Homologous recombination ,Molecular Biology ,DNA - Abstract
DNA double-strand breaks (DSB) represent a major disruption in the integrity of the genome. DSB can be generated when a replication fork encounters a DNA lesion. Recombinational repair is known to resolve such replication fork-associated DSB, but the molecular mechanism of this repair process is poorly understood in mammalian cells. In the present study, we investigated the molecular mechanism by which recombination resolves camptothecin (CPT)-induced DSB at DNA replication forks. The frequency of homologous recombination (HR) was measured using V79/SPD8 cells which contain a duplication in the endogenous hprt gene that is resolved by HR. We demonstrate that DSB associated with replication forks induce HR at the hprt gene in early S phase. Further analysis revealed that these HR events involve an exchange mechanism. Both the irs1SF and V3-3 cell lines, which are deficient in HR and non-homologous end joining (NHEJ), respectively, were found to be more sensitive than wild-type cells to DSB associated with replication forks. The irs1SF cell line was more sensitive in this respect than V3-3 cells, an observation consistent with the hypothesis that DSB associated with replication forks are repaired primarily by HR. The frequency of formation of DSB associated with replication forks was not affected in HR and NHEJ deficient cells, indicating that the loss of repair, rather than the formation of DSB associated with replication forks is responsible for the increased sensitivity of the mutant strains. We propose that the presence of DSB associated with replication forks rapidly induces HR via an exchange mechanism and that HR plays a more prominent role in the repair of such DSB than does NHEJ.
- Published
- 2001
- Full Text
- View/download PDF
29. Session 1: Signal Transduction
- Author
-
Thomas Helleday
- Subjects
Nuclear receptor ,Biochemistry ,G protein ,Signal transducing adaptor protein ,General Medicine ,Biology ,Signal transduction ,Toxicology ,Receptor ,Tyrosine kinase ,Intracellular ,Protein kinase C - Abstract
There are many intracellular pathways for signal transduction that have been identified, and at this session the main focus was on different receptors, that is, intracellular, 7TM G-protein-linked, tyrosine kinases and death receptors. Other areas covered were c-fos and signal transduction in vivo, substrates that interact with C-kinase and effects of bacterial toxins on small GTP-binding proteins.
- Published
- 1998
- Full Text
- View/download PDF
30. Introducing a new method for absolute quantification of DNA repair proteins in relation to drug development: LC–MS/MS with isotope dilution
- Author
-
Pawel Jaruga, Thomas Helleday, Ann-Sofie Jemth, Alessandro Tona, S.D. Leona, Olga Loseva, Mark S. Lowenthal, Prasad T. Reddy, Miral Dizdaroglu, and Erdem Coskun
- Subjects
Label-free quantification ,Chromatography ,Drug development ,Chemistry ,DNA repair ,Absolute quantification ,Lc ms ms ,General Medicine ,Isotope dilution ,Toxicology - Published
- 2016
- Full Text
- View/download PDF
31. Ex vivo culture of circulating tumour cell derived explants to facilitate rapid therapy testing in small cell lung cancer
- Author
-
Thomas Helleday, D. Potter, Kristopher K. Frese, Alice Lallo, U. Warpman Berglund, and Caroline Dive
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Cell ,Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Non small cell ,Ex vivo ,Explant culture - Published
- 2016
- Full Text
- View/download PDF
32. MTH1 inhibitors in cancer treatment
- Author
-
Thomas Helleday
- Subjects
DNA damage ,DNA repair ,DNA replication ,Cancer ,Hematology ,DNA Repair Pathway ,Biology ,medicine.disease ,medicine.disease_cause ,Cell biology ,Oncology ,Cancer cell ,medicine ,DNA mismatch repair ,Oxidative stress - Abstract
DNA damaging agents, i.e., radio- and chemotherapy, constitute the backbone for treatment of a wide variety of cancers and may result in complete cure from the disease. Emerging data demonstrate that cancer cells harbour high level of endogenous DNA damage caused by replication, oxidative and other DNA damage stress. Consequently, cancer cells may require a specific DNA repair pathway to mediate survival to the high load of endogenous DNA damage. Cancers have deregulated levels of reactive oxygen species (ROS), damaging both DNA and free dNTPs. The MTH1 protein sanitises oxidized dNTP pools, converting 8-oxo-dGTP to 8-oxo-dGMP, to prevent incorporation of damaged bases during DNA replication. MTH1 overexpression reverses the mutator phenotype caused by mismatch repair defects and prevents Ras-induced senescence by suppressing the overall level of DNA damage. These data suggest that a majority of damage in cancer cells occur on the free dNTP pool and that this need sanitation for cancer cell survival. Here we show that cancer cells are dependent on MTH1 activity for survival, due to the effects of MTH1 in preventing incorporation of oxidized dNTPs into DNA to avoid ATM and p53 mediated apoptosis. As MTH1-/- mice are viable and MTH1 is not required for survival of non-transformed cells, targeting MTH1 may selectively cause DNA damage to cancer cells. We validate MTH1 as an anti-cancer target in vivo and describe small molecules, TH287 and TH588 that potently and selectively inhibit MTH1. Protein co-crystal structures demonstrate that the compounds bind as inhibitors in the enzymatic pocket of MTH1. These first-in-class inhibitors of the Nudix hydrolase family cause increased incorporation of oxidized dNTPs in cells subject to high ROS levels, causing DNA damage and cytotoxicity to cancer cells. This study exemplifies a new general therapeutic approach to convert oxidative stress to cytotoxic DNA damage and cancer cell death.
- Published
- 2015
- Full Text
- View/download PDF
33. P047 Castration promotes radiosensitivity by direct regulation of DNA repair in prostate cancer
- Author
-
Thomas Helleday, H. Hamberg, Niklas Schultz, Firas L.T. Al-Ubaidi, and Torvald Granfors
- Subjects
medicine.medical_specialty ,business.industry ,DNA repair ,Urology ,medicine.disease ,Prostate cancer ,chemistry.chemical_compound ,Castration ,chemistry ,medicine ,Cancer research ,Radiosensitivity ,business - Published
- 2014
- Full Text
- View/download PDF
34. 240 The combination of Chk-1 and ATR inhibitor synergistically kills cancer cells
- Author
-
Niklas Schultz, U.W. Berglund, Thomas Helleday, Anna Hagenkort, Tobias Koolmeister, Oliver Mortusewicz, Kumar Sanjiv, John Pollard, Martin Scobie, Peter A. Charlton, and Philip Michael Reaper
- Subjects
Cancer Research ,Oncology ,Chemistry ,Cancer cell ,Cancer research - Published
- 2014
- Full Text
- View/download PDF
35. 6: Targeting MTH1 prevents sanitation of oxidised dNTP pools and causes cancer-selective DNA damage and cell death
- Author
-
Thomas Helleday
- Subjects
Cancer Research ,Programmed cell death ,Oncology ,Sanitation ,Biochemistry ,DNA damage ,Cancer research ,medicine ,Cancer ,Biology ,medicine.disease - Published
- 2014
- Full Text
- View/download PDF
36. 365: The interplay between the redox environment, MTH1 and cancer
- Author
-
Ulrika Warpman-Berglund, Thomas Helleday, Roland Fiskesund, and L. Braeutigam
- Subjects
Cancer Research ,Oncology ,Chemistry ,Cancer research ,medicine ,Cancer ,medicine.disease ,Redox - Published
- 2014
- Full Text
- View/download PDF
37. 835: Inhibiting MTH1 kills cancer cells by preventing sanitisation of oxidised dNTP pool
- Author
-
Jonas Nilsson, Helge Gad, Tobias Koolmeister, Therese Pham, Camilla Göktürk, Martin Scobie, Thomas Helleday, Saeed Eshtad, Ann-Sofie Jemth, and U. Warpman Berglund
- Subjects
Cancer Research ,Oncology ,Chemistry ,Cancer cell ,Cancer research - Published
- 2014
- Full Text
- View/download PDF
38. PARP-3 is a mono-ADP-ribosylase that activates PARP-1 in the absence of DNA
- Author
-
Thomas Helleday, Lari Lehtiö, Tobias Karlberg, Helen E. Bryant, Ann-Sofie Jemth, Herwig Schüler, and Olga Loseva
- Subjects
HMG-box ,DNA Repair ,DNA damage ,DNA repair ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,Cell Cycle Proteins ,Hydroxylamine ,Biochemistry ,Histones ,chemistry.chemical_compound ,Humans ,DNA Breaks, Single-Stranded ,Enzyme Inhibitors ,RNA, Small Interfering ,Replication protein A ,Molecular Biology ,chemistry.chemical_classification ,DNA ligase ,Adenosine Diphosphate Ribose ,biology ,Hydrolysis ,DNA ,Cell Biology ,DNA repair protein XRCC4 ,Molecular biology ,Proliferating cell nuclear antigen ,Protein Structure, Tertiary ,3-Iodobenzylguanidine ,DNA Topoisomerases, Type I ,chemistry ,Mercuric Chloride ,biology.protein ,Additions and Corrections ,Camptothecin ,Poly(ADP-ribose) Polymerases ,Topoisomerase I Inhibitors ,Nucleotide excision repair - Abstract
The PARP-3 protein is closely related to the PARP-1 and PARP-2 proteins, which are involved in DNA repair and genome maintenance. Here, we characterized the biochemical properties of human PARP-3. PARP-3 is able to ADP-ribosylate itself as well as histone H1, a previously unknown substrate for PARP-3. PARP-3 is not activated upon binding to DNA and is a mono-ADP-ribosylase, in contrast to PARP-1 and PARP-2. PARP-3 interacts with PARP-1 and activates PARP-1 in the absence of DNA, resulting in synthesis of polymers of ADP-ribose. The N-terminal WGR domain of PARP-3 is involved in this activation. The functional interaction between PARP-3 and PARP-1 suggests that it may have a role in DNA repair. However, here we report that PARP-3 small interfering RNA-depleted cells are not sensitive to the topoisomerase I poison camptothecin, inducing DNA single-strand breaks, and repair these lesions as efficiently as wild-type cells. Altogether, these results suggest that the interaction between PARP-1 and PARP-3 is unrelated to DNA single-strand break repair.
- Published
- 2012
- Full Text
- View/download PDF
39. DNA repair pathways as target for cancer therapy
- Author
-
Thomas Helleday
- Subjects
DNA repair ,Cancer research ,Cancer therapy ,General Medicine ,Biology ,Toxicology - Published
- 2012
- Full Text
- View/download PDF
40. 247 INVITED PARP Inhibitors Sensitivity and Resistance
- Author
-
Thomas Helleday
- Subjects
Cancer Research ,Oncology ,Chemistry ,Poly ADP ribose polymerase ,Cancer research ,Sensitivity (control systems) - Published
- 2011
- Full Text
- View/download PDF
41. Anti-proliferative effects of taurine conjugated fatty acids
- Author
-
Thomas Helleday, Katharina Slätis, Vicky Chatzakos, Mary C. Hunt, and Tatjana Djureinovic
- Subjects
chemistry.chemical_classification ,Taurine ,Organic Chemistry ,Fatty acid ,Cell Biology ,Conjugated system ,Anti proliferative ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Free fatty acid receptor ,Molecular Biology ,Polyunsaturated fatty acid - Published
- 2010
- Full Text
- View/download PDF
42. 611 Homologous recombination in cancer development, treatment and development of drug resistance
- Author
-
Thomas Helleday
- Subjects
Cancer Research ,Oncology ,Cancer research ,Drug resistance ,Cancer development ,Biology ,Homologous recombination - Published
- 2010
- Full Text
- View/download PDF
43. 156 Exploiting cancer defects in targeted therapy
- Author
-
Thomas Helleday
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine.medical_treatment ,medicine ,Cancer ,business ,medicine.disease ,Bioinformatics ,Targeted therapy - Published
- 2009
- Full Text
- View/download PDF
44. Anti-proliferative effects of glycine conjugated fatty acids
- Author
-
Vicky Chatzakos, Tatjana Djureinovic, Thomas Helleday, and Mary Hunt
- Subjects
Organic Chemistry ,Cell Biology ,Molecular Biology ,Biochemistry - Published
- 2009
- Full Text
- View/download PDF
45. 119 EACR award DNA replication repair: molecular insights towards new approaches to targeted anti-cancer therapy
- Author
-
Thomas Helleday
- Subjects
Genetics ,Cancer Research ,Oncology ,DNA replication ,Cancer therapy ,Computational biology ,Biology - Published
- 2007
- Full Text
- View/download PDF
46. P-128 Regulation of RAD51 levels in response to etoposide (VP16) induced DNA strand breaks in small cell lung cancer cells
- Author
-
Mogens Spang-Thomsen, Thomas Helleday, Lasse Tengbjerg Hansen, and Claus Storgaard Sørensen
- Subjects
Pulmonary and Respiratory Medicine ,Cancer Research ,business.industry ,RAD51 ,chemistry.chemical_compound ,Oncology ,chemistry ,medicine ,Cancer research ,Non small cell ,business ,Etoposide ,DNA ,medicine.drug - Published
- 2003
- Full Text
- View/download PDF
47. P XVII B.13 Interaction of fatty acids in mutagenicity
- Author
-
Thomas Helleday, Dag Jenssen, and Eva Mârtensson
- Subjects
Health, Toxicology and Mutagenesis ,Genetics ,Molecular Biology - Published
- 1997
- Full Text
- View/download PDF
48. P VIII.8 Recombinogenic effects may diverge depending on different recombination mechanisms
- Author
-
Thomas Helleday and Dag Jenssen
- Subjects
Chemistry ,Health, Toxicology and Mutagenesis ,Genetics ,Biophysics ,Molecular Biology ,Recombination - Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.