723 results on '"10061 Institute of Molecular Cancer Research"'
Search Results
2. Direct visualization of transcription-replication conflicts reveals post-replicative DNA:RNA hybrids
- Author
-
Stoy, Henriette, Zwicky, Katharina, Kuster, Danina, Lang, Kevin S, Krietsch, Jana, Crossley, Magdalena P, Schmid, Jonas A, Cimprich, Karlene A, Merrikh, Houra, Lopes, Massimo, University of Zurich, and Lopes, Massimo
- Subjects
1315 Structural Biology ,Structural Biology ,10061 Institute of Molecular Cancer Research ,1312 Molecular Biology ,570 Life sciences ,biology ,610 Medicine & health ,Molecular Biology - Abstract
Transcription-replication collisions (TRCs) are crucial determinants of genome instability. R-loops were linked to head-on TRCs and proposed to obstruct replication fork progression. The underlying mechanisms, however, remained elusive due to the lack of direct visualization and of non-ambiguous research tools. Here, we ascertained the stability of estrogen-induced R-loops on the human genome, visualized them directly by electron microscopy (EM), and measured R-loop frequency and size at the single-molecule level. Combining EM and immuno-labeling on locus-specific head-on TRCs in bacteria, we observed the frequent accumulation of DNA:RNA hybrids behind replication forks. These post-replicative structures are linked to fork slowing and reversal across conflict regions and are distinct from physiological DNA:RNA hybrids at Okazaki fragments. Comet assays on nascent DNA revealed a marked delay in nascent DNA maturation in multiple conditions previously linked to R-loop accumulation. Altogether, our findings suggest that TRC-associated replication interference entails transactions that follow initial R-loop bypass by the replication fork.
- Published
- 2023
- Full Text
- View/download PDF
3. PPM1D activity promotes the replication stress caused by cyclin E1 overexpression
- Author
-
Martinikova, Andra S, Stoyanov, Miroslav, Oravetzova, Anna, Kok, Yannick P, Yu, Shibo, Dobrovolna, Jana, Janscak, Pavel, van Vugt, Marcel, Macurek, Libor, and University of Zurich
- Subjects
10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health - Published
- 2023
4. Excessive reactive oxygen species induce transcription-dependent replication stress
- Author
-
Andrs, Martin, Stoy, Henriette, Boleslavska, Barbora, Chappidi, Nagaraja, Kanagaraj, Radhakrishnan, Nascakova, Zuzana, Menon, Shruti, Rao, Satyajeet, Oravetzova, Anna, Dobrovolna, Jana, Surendranath, Kalpana, Lopes, Massimo, Janscak, Pavel, University of Zurich, and Janscak, Pavel
- Subjects
1000 Multidisciplinary ,Multidisciplinary ,1300 General Biochemistry, Genetics and Molecular Biology ,10061 Institute of Molecular Cancer Research ,General Physics and Astronomy ,570 Life sciences ,biology ,610 Medicine & health ,1600 General Chemistry ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,3100 General Physics and Astronomy - Abstract
Elevated levels of reactive oxygen species (ROS) reduce replication fork velocity by causing dissociation of the TIMELESS-TIPIN complex from the replisome. Here, we show that ROS generated by exposure of human cells to the ribonucleotide reductase inhibitor hydroxyurea (HU) promote replication fork reversal in a manner dependent on active transcription and formation of co-transcriptional RNA:DNA hybrids (R-loops). The frequency of R-loop-dependent fork stalling events is also increased after TIMELESS depletion or a partial inhibition of replicative DNA polymerases by aphidicolin, suggesting that this phenomenon is due to a global replication slowdown. In contrast, replication arrest caused by HU-induced depletion of deoxynucleotides does not induce fork reversal but, if allowed to persist, leads to extensive R-loop-independent DNA breakage during S-phase. Our work reveals a link between oxidative stress and transcription-replication interference that causes genomic alterations recurrently found in human cancer.
- Published
- 2023
5. The importance of nuclear RAGE-Mcm2 axis in diabetes or cancer-associated replication stress
- Author
-
Han, Zhe, Andrs, Martin, Madhavan, Bindhu K, Kaymak, Serap, Sulaj, Alba, Kender, Zoltan, Kopf, Stefan, Kihm, Lars, Pepperkok, Rainer, Janscak, Pavel, Nawroth, Peter, Kumar, Varun, University of Zurich, Nawroth, Peter, and Kumar, Varun
- Subjects
1311 Genetics ,10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health - Published
- 2023
- Full Text
- View/download PDF
6. Treatment with Helicobacter pylori-derived VacA attenuates allergic airway disease
- Author
-
Reuter, Sebastian, Raspe, Jonas, Uebner, Hendrik, Contoyannis, Alexandros, Pastille, Eva, Westendorf, Astrid M, Caso, Georgia C, Cover, Timothy L, Müller, Anne, Taube, Christian, University of Zurich, and Reuter, Sebastian
- Subjects
2403 Immunology ,Immunology ,10061 Institute of Molecular Cancer Research ,2723 Immunology and Allergy ,Medizin ,Immunology and Allergy ,570 Life sciences ,biology ,610 Medicine & health - Abstract
BackgroundAsthma is an incurable heterogeneous disease with variations in clinical and underlying immunological phenotype. New approaches could help to support existing therapy concepts. Neonatal infection of mice with Helicobacter pylori or administration of H. pylori-derived extracts or molecules after birth have been shown to prevent the development of allergic airway disease later in life. This study evaluated the potential therapeutic efficacy of H. pylori vacuolating cytotoxin A (VacA) in allergic airway inflammation and investigated the underlying immunological mechanisms for its actions.MethodsMurine models of allergic airway diseases, and murine and human in vitro models were used.ResultsIn both an acute model and a therapeutic house dust mite model of allergic airway disease, treatment with H. pylori-derived VacA reduced several asthma hallmarks, including airway hyperresponsiveness, inflammation and goblet cell metaplasia. Flow cytometry and ELISA analyses revealed induction of tolerogenic dendritic cells (DC) and FoxP3 positive regulatory T cells (Tregs), and a shift in the composition of allergen-specific immunoglobulins. Depletion of Tregs during treatment with VacA reversed treatment-mediated suppression of allergic airway disease. Human monocyte derived DCs (moDC) that were exposed to VacA induced Tregs in co-cultured naïve autologous T cells, replicating key observations made in vivo.ConclusionH. pylori-derived VacA suppressed allergic airway inflammation via induction of Tregs in both allergic airway disease models. These data suggest that the immunomodulatory activity of VacA could potentially be exploited for the prevention and treatment of allergic airway disease.
- Published
- 2023
7. Functional Analysis Identifies Damaging CHEK2 Missense Variants Associated with Increased Cancer Risk
- Author
-
Boonen, Rick A C M, Wiegant, Wouter W, Celosse, Nandi, Vroling, Bas, Heijl, Stephan, Kote-Jarai, Zsofia, Mijuskovic, Martina, Cristea, Simona, Solleveld-Westerink, Nienke, van Wezel, Tom, Beerenwinkel, Niko, Eeles, Rosalind, Devilee, Peter, Vreeswijk, Maaike P G, Marra, Giancarlo, van Attikum, Haico, University of Zurich, and van Attikum, Haico
- Subjects
Male ,Mice, Knockout ,Cancer Research ,Mice, 129 Strain ,10061 Institute of Molecular Cancer Research ,Mutation, Missense ,Prostatic Neoplasms ,610 Medicine & health ,Breast Neoplasms ,Pedigree ,Checkpoint Kinase 2 ,Oncology ,Risk Factors ,Neoplasms ,570 Life sciences ,biology ,Animals ,Humans ,2730 Oncology ,1306 Cancer Research ,Female ,Genetic Predisposition to Disease ,skin and connective tissue diseases ,Cells, Cultured - Abstract
Heterozygous carriers of germline loss-of-function variants in the tumor suppressor gene checkpoint kinase 2 (CHEK2) are at an increased risk for developing breast and other cancers. While truncating variants in CHEK2 are known to be pathogenic, the interpretation of missense variants of uncertain significance (VUS) is challenging. Consequently, many VUS remain unclassified both functionally and clinically. Here we describe a mouse embryonic stem (mES) cell-based system to quantitatively determine the functional impact of 50 missense VUS in human CHEK2. By assessing the activity of human CHK2 to phosphorylate one of its main targets, Kap1, in Chek2 knockout mES cells, 31 missense VUS in CHEK2 impaired protein function to a similar extent as truncating variants, and 9 CHEK2 missense VUS resulted in intermediate functional defects. Mechanistically, most VUS impaired CHK2 kinase function by causing protein instability or by impairing activation through (auto)phosphorylation. Quantitative results showed that the degree of CHK2 kinase dysfunction correlates with an increased risk for breast cancer. Both damaging CHEK2 variants as a group (OR 2,23; 95% CI 1,62-3,07; pG/p.D162G, was also identified, which co-segregated with familial prostate cancer. Altogether, these functional assays efficiently and reliably identified VUS in CHEK2 that associate with cancer., Cancer Research, 82 (4), ISSN:0008-5472, ISSN:1538-7445
- Published
- 2021
- Full Text
- View/download PDF
8. DDX17 helicase promotes resolution of R-loop-mediated transcription-replication conflicts in human cells
- Author
-
Boleslavska, Barbora, Oravetzova, Anna, Shukla, Kaustubh, Nascakova, Zuzana, Ibini, Oluwakemi Ngozi, Hasanova, Zdenka, Andrs, Martin, Kanagaraj, Radhakrishnan, Dobrovolna, Jana, Janscak, Pavel, and University of Zurich
- Subjects
10061 Institute of Molecular Cancer Research ,Genetics ,570 Life sciences ,biology ,610 Medicine & health - Abstract
R-loops are three-stranded nucleic acid structures composed of an RNA:DNA hybrid and displaced DNA strand. These structures can halt DNA replication when formed co-transcriptionally in the opposite orientation to replication fork progression. A recent study has shown that replication forks stalled by co-transcriptional R-loops can be restarted by a mechanism involving fork cleavage by MUS81 endonuclease, followed by ELL-dependent reactivation of transcription, and fork religation by the DNA ligase IV (LIG4)/XRCC4 complex. However, how R-loops are eliminated to allow the sequential restart of transcription and replication in this pathway remains elusive. Here, we identified the human DDX17 helicase as a factor that associates with R-loops and counteracts R-loop-mediated replication stress to preserve genome stability. We show that DDX17 unwinds R-loops in vitro and promotes MUS81-dependent restart of R-loop-stalled forks in human cells in a manner dependent on its helicase activity. Loss of DDX17 helicase induces accumulation of R-loops and the formation of R-loop-dependent anaphase bridges and micronuclei. These findings establish DDX17 as a component of the MUS81–LIG4–ELL pathway for resolution of R-loop-mediated transcription–replication conflicts, which may be involved in R-loop unwinding.
- Published
- 2022
9. Stress-triggered hematopoietic stem cell proliferation relies on PrimPol-mediated repriming
- Author
-
Kurt Jacobs, Cyril Doerdelmann, Jana Krietsch, Daniel González-Acosta, Nicolas Mathis, Saul Kushinsky, Estrella Guarino, Carmen Gómez-Escolar, Dolores Martinez, Jonas A. Schmid, Peter J. Leary, Raimundo Freire, Almudena R. Ramiro, Christine M. Eischen, Juan Mendez, Massimo Lopes, European Research Council, Ministerio de Ciencia e Innovación (España), Agencia Estatal de Investigación (España), University of Zurich, Mendez, Juan, and Lopes, Massimo
- Subjects
DNA Replication ,10061 Institute of Molecular Cancer Research ,610 Medicine & health ,Cell Biology ,Hematopoietic Stem Cells ,Article ,Hematopoiesis ,1307 Cell Biology ,Mice ,1312 Molecular Biology ,570 Life sciences ,biology ,Animals ,Molecular Biology ,DNA Damage ,Cell Proliferation - Abstract
Stem cell division is linked to tumorigenesis by yet-elusive mechanisms. The hematopoietic system reacts to stress by triggering hematopoietic stem and progenitor cell (HSPC) proliferation, which can be accompanied by chromosomal breakage in activated hematopoietic stem cells (HSCs). However, whether these lesions persist in their downstream progeny and induce a canonical DNA damage response (DDR) remains unclear. Inducing HSPC proliferation by simulated viral infection, we report that the associated DNA damage is restricted to HSCs and that proliferating HSCs rewire their DDR upon endogenous and clastogen-induced damage. Combining transcriptomics, single-cell and single-molecule assays on murine bone marrow cells, we found accelerated fork progression in stimulated HSPCs, reflecting engagement of PrimPol-dependent repriming, at the expense of replication fork reversal. Ultimately, competitive bone marrow transplantation revealed the requirement of PrimPol for efficient HSC amplification and bone marrow reconstitution. Hence, fine-tuning replication fork plasticity is essential to support stem cell functionality upon proliferation stimuli. Sí
- Published
- 2022
10. DNA-PKcs promotes fork reversal and chemoresistance
- Author
-
Dibitetto, Diego, Marshall, Shannon, Sanchi, Andrea, Liptay, Martin, Badar, Jumana, Lopes, Massimo, Rottenberg, Sven, Smolka, Marcus B., University of Zurich, Dibitetto, Diego, and Smolka, Marcus B
- Subjects
DNA Replication ,DNA End-Joining Repair ,DNA Repair ,10061 Institute of Molecular Cancer Research ,610 Medicine & health ,Cell Biology ,DNA ,1307 Cell Biology ,Drug Resistance, Neoplasm ,1312 Molecular Biology ,570 Life sciences ,biology ,630 Landwirtschaft ,DNA Breaks, Double-Stranded ,Molecular Biology ,DNA Damage - Abstract
The DNA-PKcs kinase mediates the repair of DNA double-strand breaks via classical non-homologous end joining (NHEJ). DNA-PKcs is also recruited to active replication forks, although a role for DNA-PKcs in the control of fork dynamics is unclear. Here, we identify a crucial role for DNA-PKcs in promoting fork reversal, a process that stabilizes stressed replication forks and protects genome integrity. DNA-PKcs promotes fork reversal and slowing in response to several replication stress-inducing agents in a manner independent of its role in NHEJ. Cells lacking DNA-PKcs activity show increased DNA damage during S-phase and cellular sensitivity to replication stress. Notably, prevention of fork slowing and reversal via DNA-PKcs inhibition efficiently restores chemotherapy sensitivity in BRCA2-deficient mammary tumors with acquired PARPi resistance. Together, our data uncover a new key regulator of fork reversal and show how DNA-PKcs signaling can be manipulated to alter fork dynamics and drug resistance in cancer.
- Published
- 2022
- Full Text
- View/download PDF
11. Molecular Characterization of Circulating Tumor DNA in Pediatric Rhabdomyosarcoma: A Feasibility Study
- Author
-
Ruhen, Olivia, Lak, Nathalie S M, Stutterheim, Janine, Danielli, Sara G, et al, Surdez, Didier, Schäfer, Beat W, and University of Zurich
- Subjects
10036 Medical Clinic ,10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health ,10046 Balgrist University Hospital, Swiss Spinal Cord Injury Center - Published
- 2022
12. FAN1, a DNA Repair Nuclease, as a Modifier of Repeat Expansion Disorders
- Author
-
Gagan B. Panigrahi, Jean-Yves Masson, Amit Laxmikant Deshmukh, Antonio Porro, Mohiuddin Mohiuddin, Stella Lanni, Christopher E. Pearson, Alessandro A. Sartori, Marie-Christine Caron, University of Zurich, Jones, Lesley, Pearson, Christopher E, and Wheeler, Vanessa
- Subjects
0301 basic medicine ,DNA Repair ,DNA repair ,2804 Cellular and Molecular Neuroscience ,Clinical Neurology ,610 Medicine & health ,Review ,Biology ,Genomic Instability ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Huntington's disease ,FAN1 ,medicine ,Animals ,Humans ,Spinocerebellar Ataxias ,Copy-number variation ,nuclease ,Gene ,repeat instability ,Genetics ,modifier ,Endodeoxyribonucleases ,Genes, Modifier ,10061 Institute of Molecular Cancer Research ,medicine.disease ,Multifunctional Enzymes ,FMR1 ,karyomegalic interstitial nephritis ,Exodeoxyribonucleases ,Huntington Disease ,2728 Neurology (clinical) ,030104 developmental biology ,Spinocerebellar ataxia ,570 Life sciences ,biology ,Neurology (clinical) ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Huntington’s disease - Abstract
FAN1 encodes a DNA repair nuclease. Genetic deficiencies, copy number variants, and single nucleotide variants of FAN1 have been linked to karyomegalic interstitial nephritis, 15q13.3 microdeletion/microduplication syndrome (autism, schizophrenia, and epilepsy), cancer, and most recently repeat expansion diseases. For seven CAG repeat expansion diseases (Huntington’s disease (HD) and certain spinocerebellar ataxias), modification of age of onset is linked to variants of specific DNA repair proteins. FAN1 variants are the strongest modifiers. Non-coding disease-delaying FAN1 variants and coding disease-hastening variants (p.R507H and p.R377W) are known, where the former may lead to increased FAN1 levels and the latter have unknown effects upon FAN1 functions. Current thoughts are that ongoing repeat expansions in disease-vulnerable tissues, as individuals age, promote disease onset. Fan1 is required to suppress against high levels of ongoing somatic CAG and CGG repeat expansions in tissues of HD and FMR1 transgenic mice respectively, in addition to participating in DNA interstrand crosslink repair. FAN1 is also a modifier of autism, schizophrenia, and epilepsy. Coupled with the association of these diseases with repeat expansions, this suggests a common mechanism, by which FAN1 modifies repeat diseases. Yet how any of the FAN1 variants modify disease is unknown. Here, we review FAN1 variants, associated clinical effects, protein structure, and the enzyme’s attributed functional roles. We highlight how variants may alter its activities in DNA damage response and/or repeat instability. A thorough awareness of the FAN1 gene and FAN1 protein functions will reveal if and how it may be targeted for clinical benefit.
- Published
- 2021
- Full Text
- View/download PDF
13. Disentangling tumorigenesis-associated DNA methylation changes in colorectal tissues from those associated with ageing
- Author
-
Mark D. Robinson, Hannah R Parker, Matthias Sauter, Federico Buffoli, Fabrizio Cereatti, Giancarlo Marra, Stephany Orjuela, Sija Sajibu, University of Zurich, and Marra, Giancarlo
- Subjects
Aging ,Cancer Research ,Carcinogenesis ,Bisulfite sequencing ,Colorectal adenoma ,Biology ,medicine.disease_cause ,Biomarkers, Tumor ,medicine ,1312 Molecular Biology ,Humans ,1306 Cancer Research ,Promoter Regions, Genetic ,Molecular Biology ,Epigenomics ,10061 Institute of Molecular Cancer Research ,Methylation ,DNA Methylation ,medicine.disease ,10124 Institute of Molecular Life Sciences ,Chromatin ,Gene Expression Regulation, Neoplastic ,CpG site ,DNA methylation ,Cancer research ,570 Life sciences ,biology ,CpG Islands ,Female ,Colorectal Neoplasms ,Research Paper - Abstract
Physiological ageing and tumorigenesis are both associated with epigenomic alterations in human tissue cells, the most extensively investigated of which entails de novo cytosine methylation (i.e., hypermethylation) within the CpG dinucleotides of CpG islands. Genomic regions that become hypermethylated during tumorigenesis are generally believed to overlap regions that acquire methylation in normal tissues as an effect of ageing. To define the extension of this overlap, we analysed the DNA methylomes of 48 large-bowel tissue samples taken from women of different ages during screening colonoscopy: 18 paired samples of normal and lesional tissues from donors harbouring a precancerous lesion and 12 samples of normal mucosa from tumour-free donors. Each sample was subjected to targeted, genome-wide bisulphite sequencing of ~2.5% of the genome, including all CpG islands. In terms of both its magnitude and extension along the chromatin, tumour-associated DNA hypermethylation in these regions was much more conspicuous than that observed in the normal mucosal samples from older (vs. younger) tumour-free donors. 83% of the ageing-associated hypermethylated regions (n = 2501) coincided with hypermethylated regions observed in tumour samples. However, 86% of the regions displaying hypermethylation in precancerous lesions (n = 16,772) showed no methylation changes in the ageing normal mucosa. The tumour-specificity of this latter hypermethylation was validated using published sets of data on DNA methylation in normal and neoplastic colon tissues. This extensive set of genomic regions displaying tumour-specific hypermethylation represents a rich vein of putative biomarkers for the early, non-invasive detection of colorectal tumours in women of all ages.
- Published
- 2022
- Full Text
- View/download PDF
14. MutSβ regulates G4-associated telomeric R-loops to maintain telomere integrity in ALT cancer cells
- Author
-
Despoina Sakellariou, Sara Thornby Bak, Esin Isik, Sonia I. Barroso, Antonio Porro, Andrés Aguilera, Jiri Bartek, Pavel Janscak, Javier Peña-Diaz, Danish Council for Independent Research, Danish Cancer Society Research Center, Nordea Foundation, Swiss National Science Foundation, Czech Science Foundation, Novo Nordisk Foundation, Swedish Research Council, Swedish Cancer Society, European Research Council, European Commission, Agencia Estatal de Investigación (España), Ministerio de Ciencia, Innovación y Universidades (España), University of Zurich, Janscak, Pavel, Peña-Diaz, Javier, and Universidad de Sevilla. Departamento de Genética
- Subjects
Cancer [CP] ,Molecular biology [CP] ,G-quadruplex ,C-circle ,10061 Institute of Molecular Cancer Research ,R-looptelomere ,Telomere Homeostasis ,610 Medicine & health ,ALT cancers ,DNA ,Telomere ,General Biochemistry, Genetics and Molecular Biology ,Mismatch repair ,1300 General Biochemistry, Genetics and Molecular Biology ,Neoplasms ,570 Life sciences ,biology ,Humans ,R-loop ,RNA, Long Noncoding ,R-Loop Structures ,C-circlemismatch repair - Abstract
Up to 15% of human cancers maintain their telomeres through a telomerase-independent mechanism, termed “alternative lengthening of telomeres” (ALT) that relies on homologous recombination between telomeric sequences. Emerging evidence suggests that the recombinogenic nature of ALT telomeres results from the formation of RNA:DNA hybrids (R-loops) between telomeric DNA and the long-noncoding telomeric repeat-containing RNA (TERRA). Here, we show that the mismatch repair protein MutSβ, a heterodimer of MSH2 and MSH3 subunits, is enriched at telomeres in ALT cancer cells, where it prevents the accumulation of telomeric G-quadruplex (G4) structures and R-loops. Cells depleted of MSH3 display increased incidence of R-loop-dependent telomere fragility and accumulation of telomeric C-circles. We also demonstrate that purified MutSβ recognizes and destabilizes G4 structures in vitro. These data suggest that MutSβ destabilizes G4 structures in ALT telomeres to regulate TERRA R-loops, which is a prerequisite for maintenance of telomere integrity during ALT., J.P.D. was supported by the Danish Council for Independent Research (DFF-4004-00117), the Danish Cancer Society (R72-A4181-13-S2), the Nordea Foundation, Agnes and Poul Friis Fond, and Magda Sofie and Aase Lutz Mindelegat. P.J. was supported by the Swiss National Science Foundation (310030_184716) and Czech Science Foundation (19-07674S). J.B. was supported by the Danish Council for Independent Research (DFF-7016-00313), the Danish Cancer Society (R204-A12617), the Novo Nordisk Foundation (200C0060590), the Czech Science Foundation (project 19-21325S), the Swedish Research Council (VR-MH 2014-46602-117891-30), and the Swedish CancerFonden (no. 170176) and A.A. by the European Research Council (ERC2014 AdG669898 TARLOOP), the Spanish Ministry of Science and Innovation (PID2019-104270GB-I00/BMC), and the European Union (FEDER).
- Published
- 2022
- Full Text
- View/download PDF
15. The ALPK1/TIFA/NF-κB axis links a bacterial carcinogen to R-loop-induced replication stress
- Author
-
Bauer, Michael, Nascakova, Zuzana, Mihai, Anca-Irina, Cheng, Phil F, Levesque, Mitchell P, Lampart, Simon, Hurwitz, Robert, Pfannkuch, Lennart, Dobrovolna, Jana, Jacobs, Melanie, Bartfeld, Sina, Dohlman, Anders, Shen, Xiling, Gall, Alevtina A, Salama, Nina R, Töpfer, Antonia, Weber, Achim, Meyer, Thomas F, Janscak, Pavel, Müller, Anne, University of Zurich, Janscak, Pavel, and Müller, Anne
- Subjects
DNA Replication ,Lipopolysaccharides ,Science ,610 Medicine & health ,1600 General Chemistry ,Microbiology ,Article ,Helicobacter Infections ,Bacterial Proteins ,1300 General Biochemistry, Genetics and Molecular Biology ,Stomach Neoplasms ,10049 Institute of Pathology and Molecular Pathology ,Cell Line, Tumor ,Humans ,lcsh:Science ,Adaptor Proteins, Signal Transducing ,Cancer ,Helicobacter pylori ,Molecular medicine ,10061 Institute of Molecular Cancer Research ,NF-kappa B ,Gastroenterology ,Glycosyltransferases ,DNA ,3100 General Physics and Astronomy ,Computational biology and bioinformatics ,Host-Pathogen Interactions ,Mutation ,570 Life sciences ,biology ,lcsh:Q ,Floxuridine ,Reactive Oxygen Species ,Protein Kinases ,DNA Damage - Abstract
Exposure of gastric epithelial cells to the bacterial carcinogen Helicobacter pylori causes DNA double strand breaks. Here, we show that H. pylori-induced DNA damage occurs co-transcriptionally in S-phase cells that activate NF-κB signaling upon innate immune recognition of the lipopolysaccharide biosynthetic intermediate β-ADP-heptose by the ALPK1/TIFA signaling pathway. DNA damage depends on the bi-functional RfaE enzyme and the Cag pathogenicity island of H. pylori, is accompanied by replication fork stalling and can be observed also in primary cells derived from gastric organoids. Importantly, H. pylori-induced replication stress and DNA damage depend on the presence of co-transcriptional RNA/DNA hybrids (R-loops) that form in infected cells during S-phase as a consequence of β-ADP-heptose/ ALPK1/TIFA/NF-κB signaling. H. pylori resides in close proximity to S-phase cells in the gastric mucosa of gastritis patients. Taken together, our results link bacterial infection and NF-κB-driven innate immune responses to R-loop-dependent replication stress and DNA damage., The bacterial pathogen Helicobacter pylori is known for its ability to induce DNA double-strand breaks in the genome of its target cells. Here, we show that H. pylori-induced DNA damage and replication stress occurs in S-phase cells as a result of R-loop-mediated transcription/replication conflicts that are triggered by activation of the ALPK1/TIFA/NF-κB signaling axis.
- Published
- 2020
- Full Text
- View/download PDF
16. The plasticity of DNA replication forks in response to clinically relevant genotoxic stress
- Author
-
Massimo Lopes, David Cortez, Matteo Berti, and University of Zurich
- Subjects
DNA Replication ,Carcinogenesis ,DNA damage ,610 Medicine & health ,Replication Origin ,Genotoxic Stress ,Biology ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Innate immune system ,10061 Institute of Molecular Cancer Research ,DNA replication ,DNA ,Cell Biology ,Chromatin ,Immunity, Innate ,Cell biology ,Crosstalk (biology) ,chemistry ,570 Life sciences ,biology ,030217 neurology & neurosurgery ,DNA Damage - Abstract
Complete and accurate DNA replication requires the progression of replication forks through DNA damage, actively transcribed regions, structured DNA and compact chromatin. Recent studies have revealed a remarkable plasticity of the replication process in dealing with these obstacles, which includes modulation of replication origin firing, of the architecture of replication forks, and of the functional organization of the replication machinery in response to replication stress. However, these specialized mechanisms also expose cells to potentially dangerous transactions while replicating DNA. In this Review, we discuss how replication forks are actively stalled, remodelled, processed, protected and restarted in response to specific types of stress. We also discuss adaptations of the replication machinery and the role of chromatin modifications during these transactions. Finally, we discuss interesting recent data on the relevance of replication fork plasticity to human health, covering its role in tumorigenesis, its crosstalk with innate immunity responses and its potential as an effective cancer therapy target.
- Published
- 2020
- Full Text
- View/download PDF
17. TGF-β production by eosinophils drives the expansion of peripherally induced neuropilin- RORγt+ regulatory T-cells during bacterial and allergen challenge
- Author
-
Fallegger, Angela, Priola, Martina, Artola-Borán, Mariela, Núñez, Nicolás Gonzalo, Wild, Sebastian, Gurtner, Alessandra, Becher, Burkhard, Yousefi, Shida, Simon, Hans-Uwe, Arnold, Isabelle C, Müller, Anne, University of Zurich, Arnold, Isabelle C, and Müller, Anne
- Subjects
2403 Immunology ,10061 Institute of Molecular Cancer Research ,Immunology ,2723 Immunology and Allergy ,570 Life sciences ,biology ,Immunology and Allergy ,610 Medicine & health ,10263 Institute of Experimental Immunology - Published
- 2022
- Full Text
- View/download PDF
18. RNAi Screening Uncovers a Synthetic Sick Interaction between CtIP and the BARD1 Tumor Suppressor
- Author
-
Bolck, Hella A, Przetocka, Sara, Meier, Roger, von Aesch, Christine, Zurfluh, Christina, Hänggi, Kay, Spegg, Vincent, Altmeyer, Matthias, Stebler, Michael, Nørrelykke, Simon F, Horvath, Peter, Sartori, Alessandro A, Porro, Antonio, Institute for Molecular Medicine Finland, University of Zurich, Sartori, Alessandro A, and Porro, Antonio
- Subjects
CHROMATIN ,DNA Repair ,replication stress ,Ubiquitin-Protein Ligases ,610 Medicine & health ,DNA-DAMAGE RESPONSE ,2700 General Medicine ,END RESECTION ,CtIP ,BARD1 ,BRCA1 ,synthetic lethality ,DNA damage ,CELL-CYCLE ,Humans ,DNA Breaks, Double-Stranded ,Genes, Tumor Suppressor ,Homologous Recombination ,EARLY EMBRYONIC LETHALITY ,REPAIR ,Endodeoxyribonucleases ,STABILITY ,PARTNER ,Tumor Suppressor Proteins ,10061 Institute of Molecular Cancer Research ,General Medicine ,570 Life sciences ,biology ,REPEATS ,1182 Biochemistry, cell and molecular biology ,RNA Interference - Abstract
Human CtIP is best known for its role in DNA end resection to initiate DNA double-strand break repair by homologous recombination. Recently, CtIP has also been shown to protect reversed replication forks from nucleolytic degradation upon DNA replication stress. However, still little is known about the DNA damage response (DDR) networks that preserve genome integrity and sustain cell survival in the context of CtIP insufficiency. Here, to reveal such potential buffering relationships, we screened a DDR siRNA library in CtIP-deficient cells to identify candidate genes that induce synthetic sickness/lethality (SSL). Our analyses unveil a negative genetic interaction between CtIP and BARD1, the heterodimeric binding partner of BRCA1. We found that simultaneous disruption of CtIP and BARD1 triggers enhanced apoptosis due to persistent replication stress-induced DNA lesions giving rise to chromosomal abnormalities. Moreover, we observed that the genetic interaction between CtIP and BARD1 occurs independently of the BRCA1-BARD1 complex formation and might be, therefore, therapeutical relevant for the treatment of BRCA-defective tumors.
- Published
- 2022
- Full Text
- View/download PDF
19. Direct R-Loop Visualization on Genomic DNA by Native Automated Electron Microscopy
- Author
-
Stoy, Henriette, Luethi, Joel, Roessler, Fabienne K, Riemann, Johannes, Kaech, Andres, Lopes, Massimo, University of Zurich, Aguilera, Andrés, Ruzov, Alexey, and Lopes, Massimo
- Subjects
1311 Genetics ,10061 Institute of Molecular Cancer Research ,1312 Molecular Biology ,570 Life sciences ,biology ,610 Medicine & health - Published
- 2022
- Full Text
- View/download PDF
20. Locus-Specific Analysis of Replication Dynamics and Detection of DNA–RNA Hybrids by Immuno Electron Microscopy
- Author
-
Stoy, Henriette, Lang, Kevin S, Merrikh, Houra, Lopes, Massimo, University of Zurich, Aguilera, Andrés, Ruzov, Alexey, and Lopes, Massimo
- Subjects
DNA Replication ,Microscopy, Electron ,1311 Genetics ,10061 Institute of Molecular Cancer Research ,1312 Molecular Biology ,570 Life sciences ,biology ,RNA ,610 Medicine & health ,DNA ,Microscopy, Immunoelectron ,Article - Abstract
DNA-RNA hybrids can interfere with DNA replication, but the underlying intermediates and molecular mechanisms have remained elusive. Here, we describe a single molecule approach that allows to monitor DNA-RNA hybrids locus-specifically in the context of ongoing replication. Using restriction digestion, gel electrophoresis and gel elution, this workflow allows to efficiently isolate replication intermediates and to study replication dynamics across a specific genomic locus. Here, we applied this procedure to isolate a bacterial genomic locus carrying an inducible transcription-replication conflict. Moreover, we combined electron microscopy with S9.6-Gold immuno-labeling to detect DNA-RNA hybrids on the isolated replication intermediates. With some limitations, this approach may be adapted to locus-specific replication analyses in different organisms.
- Published
- 2022
21. Mycobacterial infection aggravates Helicobacter pylori-induced gastric preneoplastic pathology by redirection of de novo induced Treg cells
- Author
-
Artola-Borán, Mariela, Fallegger, Angela, Priola, Martina, Jeske, Rima, Waterboer, Tim, Dohlman, Anders B, Shen, Xiling, Wild, Sebastian, He, Jiazhuo, Levesque, Mitchell P, Yousefi, Shida, Simon, Hans-Uwe, Cheng, Phil F, Müller, Anne, University of Zurich, and Müller, Anne
- Subjects
Mycobacterium Infections ,Helicobacter pylori ,QH301-705.5 ,gastric cancer ,10061 Institute of Molecular Cancer Research ,610 Medicine & health ,chemical and pharmacologic phenomena ,Mycobacterium tuberculosis ,bacterial persistence ,CD8-Positive T-Lymphocytes ,bacterial infections and mycoses ,Mycobacterium bovis ,T-Lymphocytes, Regulatory ,Helicobacter Infections ,Mice, Inbred C57BL ,1300 General Biochemistry, Genetics and Molecular Biology ,570 Life sciences ,biology ,Animals ,mutual interaction of pathogenic bacteria ,mycobacterial infection ,Biology (General) ,granuloma - Abstract
Summary: The two human pathogens Helicobacter pylori and Mycobacterium tuberculosis (Mtb) co-exist in many geographical areas of the world. Here, using a co-infection model of H. pylori and the Mtb relative M. bovis bacillus Calmette-Guérin (BCG), we show that both bacteria affect the colonization and immune control of the respective other pathogen. Co-occurring M. bovis boosts gastric Th1 responses and H. pylori control and aggravates gastric immunopathology. H. pylori in the stomach compromises immune control of M. bovis in the liver and spleen. Prior antibiotic H. pylori eradication or M. bovis-specific immunization reverses the effects of H. pylori. Mechanistically, the mutual effects can be attributed to the redirection of regulatory T cells (Treg cells) to sites of M. bovis infection. Reversal of Treg cell redirection by CXCR3 blockade restores M. bovis control. In conclusion, the simultaneous presence of both pathogens exacerbates the problems associated with each individual infection alone and should possibly be factored into treatment decisions.
- Published
- 2022
- Full Text
- View/download PDF
22. Cancer cells use self-inflicted DNA breaks to evade growth limits imposed by genotoxic stress
- Author
-
Brian D. Larsen, Jan Benada, Philip Yuk Kwong Yung, Ryan A. V. Bell, George Pappas, Vaclav Urban, Johanna K. Ahlskog, Tia T. Kuo, Pavel Janscak, Lynn A. Megeney, Simon J. Elsässer, Jiri Bartek, Claus S. Sørensen, University of Zurich, and Sørensen, Claus S
- Subjects
1000 Multidisciplinary ,Multidisciplinary ,DNA Repair ,Neoplasms ,10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health ,DNA Breaks, Double-Stranded ,DNA ,Chromatin ,DNA Damage - Abstract
Genotoxic therapy such as radiation serves as a frontline cancer treatment, yet acquired resistance that leads to tumor reoccurrence is frequent. We found that cancer cells maintain viability during irradiation by reversibly increasing genome-wide DNA breaks, thereby limiting premature mitotic progression. We identify caspase-activated DNase (CAD) as the nuclease inflicting these de novo DNA lesions at defined loci, which are in proximity to chromatin-modifying CCCTC-binding factor (CTCF) sites. CAD nuclease activity is governed through phosphorylation by DNA damage response kinases, independent of caspase activity. In turn, loss of CAD activity impairs cell fate decisions, rendering cancer cells vulnerable to radiation-induced DNA double-strand breaks. Our observations highlight a cancer-selective survival adaptation, whereby tumor cells deploy regulated DNA breaks to delimit the detrimental effects of therapy-evoked DNA damage.
- Published
- 2022
- Full Text
- View/download PDF
23. Value-based pricing of drugs with multiple indications or in combinations — lessons from Europe
- Author
-
Vokinger, Kerstin N, Kesselheim, Aaron S, University of Zurich, and Vokinger, Kerstin N
- Subjects
Oncology ,10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health ,2730 Oncology - Published
- 2022
- Full Text
- View/download PDF
24. Influence of the early-life gut microbiota on the immune responses to an inhaled allergen
- Author
-
Borbet, Timothy C, Pawline, Miranda B, Zhang, Xiaozhou, Wipperman, Matthew F, Reuter, Sebastian, Maher, Timothy, Li, Jackie, Iizumi, Tadasu, Gao, Zhan, Daniele, Megan, Taube, Christian, Koralov, Sergei B, Müller, Anne, Blaser, Martin J, and University of Zurich
- Subjects
Immunology ,Pyroglyphidae ,10061 Institute of Molecular Cancer Research ,Immunity ,Medizin ,Amoxicillin ,610 Medicine & health ,Allergens ,Azithromycin ,Immunoglobulin E ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Mice ,Th2 Cells ,Immunology and Allergy ,Animals ,Cytokines ,570 Life sciences ,biology - Abstract
Antibiotics, among the most used medications in children, affect gut microbiome communities and metabolic functions. These changes in microbiota structure can impact host immunity. We hypothesized that early-life microbiome alterations would lead to increased susceptibility to allergy and asthma. To test this, mouse pups between postnatal days 5-9 were orally exposed to water (control) or to therapeutic doses of azithromycin or amoxicillin. Later in life, these mice were sensitized and challenged with a model allergen, house dust mite (HDM), or saline. Mice with early-life azithromycin exposure that were challenged with HDM had increased IgE and IL-13 production by CD4
- Published
- 2022
- Full Text
- View/download PDF
25. TARG1 protects against toxic DNA ADP-ribosylation
- Author
-
Andrea Sanchi, Giuliana Katharina Moeller, Callum Tromans-Coia, Ivan Ahel, Gyula Timinszky, Massimo Lopes, and University of Zurich
- Subjects
DNA Replication ,DNA Repair ,AcademicSubjects/SCI00010 ,Bacterial Toxins ,DNA, Single-Stranded ,610 Medicine & health ,DNA ADP-ribosylation ,Genome Integrity, Repair and Replication ,Biology ,medicine.disease_cause ,Genome ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,1311 Genetics ,Genetics ,medicine ,Humans ,030304 developmental biology ,Adenosine Diphosphate Ribose ,0303 health sciences ,Toxin ,10061 Institute of Molecular Cancer Research ,DNA ,Cell biology ,genomic DNA ,chemistry ,030220 oncology & carcinogenesis ,ADP-ribosylation ,570 Life sciences ,biology ,Thiolester Hydrolases ,Antitoxin ,Thymidine - Abstract
ADP-ribosylation is a modification that targets a variety of macromolecules and regulates a diverse array of important cellular processes. ADP-ribosylation is catalysed by ADP-ribosyltransferases and reversed by ADP-ribosylhydrolases. Recently, an ADP-ribosyltransferase toxin termed ‘DarT’ from bacteria, which is distantly related to human PARPs, was shown to modify thymidine in single-stranded DNA in a sequence specific manner. The antitoxin of DarT is the macrodomain containing ADP-ribosylhydrolase DarG, which shares striking structural homology with the human ADP-ribosylhydrolase TARG1. Here, we show that TARG1, like DarG, can reverse thymidine-linked DNA ADP-ribosylation. We find that TARG1-deficient human cells are extremely sensitive to DNA ADP-ribosylation. Furthermore, we also demonstrate the first detection of reversible ADP-ribosylation on genomic DNA in vivo from human cells. Collectively, our results elucidate the impact of DNA ADP-ribosylation in human cells and provides a molecular toolkit for future studies into this largely unknown facet of ADP-ribosylation.
- Published
- 2021
- Full Text
- View/download PDF
26. FAN1 controls mismatch repair complex assembly via MLH1 retention to stabilize CAG repeat expansion in Huntington's disease
- Author
-
Alessandro A. Sartori, Gabriel Balmus, Gillian P. Bates, Konstantinos Thalassinos, Hilary Wilkinson, Joseph Hamilton, Michael Flower, Robert Goold, Emma L. Bunting, Antonio Porro, Sarah G. Aldous, Thomas Menneteau, Jose R. Vicente, Sarah J. Tabrizi, Nicholas D. Allen, Balmus, Gabriel [0000-0003-2872-4468], Apollo - University of Cambridge Repository, University of Zurich, Balmus, Gabriel, Tabrizi, Sarah J, Menneteau, Thomas [0000-0003-1831-5425], and Tabrizi, Sarah J [0000-0003-2716-2045]
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Mismatch repair complex ,DNA repair ,Context (language use) ,610 Medicine & health ,Binding, Competitive ,DNA Mismatch Repair ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mismatch repair ,Mice ,1300 General Biochemistry, Genetics and Molecular Biology ,Fan1 ,Cell Line, Tumor ,Huntington’s Disease ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Fan1 Nuclease Activity ,Nuclease ,Huntingtin Protein ,Endodeoxyribonucleases ,biology ,FAN1 ,10061 Institute of Molecular Cancer Research ,MLH1 ,Brain ,Msh3 ,Multifunctional Enzymes ,Cell biology ,Exodeoxyribonucleases ,HEK293 Cells ,Huntington Disease ,MSH3 ,Cag Instability ,MutS Homolog 3 Protein ,biology.protein ,570 Life sciences ,Repeat Expansion ,DNA mismatch repair ,Trinucleotide repeat expansion ,MutL Protein Homolog 1 ,Trinucleotide Repeat Expansion ,DNA Damage ,Protein Binding ,Gwas - Abstract
Summary CAG repeat expansion in the HTT gene drives Huntington’s disease (HD) pathogenesis and is modulated by DNA damage repair pathways. In this context, the interaction between FAN1, a DNA-structure-specific nuclease, and MLH1, member of the DNA mismatch repair pathway (MMR), is not defined. Here, we identify a highly conserved SPYF motif at the N terminus of FAN1 that binds to MLH1. Our data support a model where FAN1 has two distinct functions to stabilize CAG repeats. On one hand, it binds MLH1 to restrict its recruitment by MSH3, thus inhibiting the assembly of a functional MMR complex that would otherwise promote CAG repeat expansion. On the other hand, it promotes accurate repair via its nuclease activity. These data highlight a potential avenue for HD therapeutics in attenuating somatic expansion., Graphical abstract, Highlights • FAN1 binds MLH1 via conserved 126SPYF129 residues, acting as a canonical MIP-box • FAN1-MLH1 binding regulates mismatch repair activity and complex formation • FAN1-MLH1 binding regulates the HTT CAG expansion rate, FAN1 modifies Huntington’s disease pathogenesis, but the mechanism has remained elusive. Goold et al. demonstrate that FAN1 binds MLH1 through residues 126SPYF129, competing with MSH3, and sequesters MLH1 from the mismatch repair pathway. In turn, this reduces mismatch repair activity and suppresses expansion of the pathogenic HTT CAG trinucleotide repeat.
- Published
- 2021
- Full Text
- View/download PDF
27. Associations With Definitive Outcomes and Clinical Benefit of Cancer Drugs at the Time of Marketing Approval and in the Postmarketing Period
- Author
-
Jose Carlos Tapia, Thomas J Hwang, Arnoud J. Templeton, Ariadna Tibau, Agustí Barnadas, Aida Bujosa, Kerstin Noëlle Vokinger, Consolación Molto, Ignasi Gich, Eitan Amir, and University of Zurich
- Subjects
medicine.medical_specialty ,business.industry ,Immune checkpoint inhibitors ,11476 Digital Society Initiative ,Cancer drugs ,10061 Institute of Molecular Cancer Research ,MEDLINE ,610 Medicine & health ,Quality of life ,Oncology ,Internal medicine ,medicine ,Overall survival ,570 Life sciences ,biology ,business ,Companion diagnostic ,Value framework - Abstract
Background: Most anticancer drugs are approved by regulatory agencies based on surrogate measures. This article explores the variables associated with overall survival (OS), quality of life (QoL), and substantial clinical benefit among anticancer drugs at the time of approval and in the postmarketing period. Methods: Anticancer drugs approved by the FDA between January 2006 and December 2015 and with postmarketing follow-up until April 2019 were identified. We evaluated trial-level data supporting approval and any updated OS and/or QoL data. We applied the ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS) and the ASCO Value Framework (ASCO-VF) to initial and follow-up studies. Results: We found that 58 drugs were approved for 96 indications based on 96 trials. At registration, approval was based on improved OS in 39 trials (41%) and improved QoL in 16 of 45 indications (36%). Postmarketing data showed an improvement in OS for 28 of 59 trials (47%) and in QoL for 22 of 48 indications (46%). At the time of approval, 25 of 94 (27%) and 26 of 80 scorable trials (33%) met substantial benefit thresholds using the ESMO-MCBS and ASCO-VF, respectively. In the postmarketing period, 37 of 69 (54%) and 35 of 65 (54%) trials met the substantial benefit thresholds. Drugs with companion diagnostics and immune checkpoint inhibitors were associated significantly with substantial clinical benefit. Conclusions: Compared with the time of approval, more anticancer drugs showed improved OS and QoL and met the ESMO-MCBS or ASCO-VF thresholds for substantial benefit over the course of postmarketing time. However, only approximately half of the trials met the threshold for substantial benefit. Companion diagnostic drugs and immunotherapy seemed to be associated with greater clinical benefit.
- Published
- 2021
28. FANCD2-associated nuclease 1 partially compensates for the lack of Exonuclease 1 in mismatch repair
- Author
-
Gene Koh, Josef Jiricny, Serena Nik-Zainal, Hiroyuki Sasanuma, Alessandro A. Sartori, Saho Kobayashi-Era, Goncalo Oliveira, Katja Kratz, Andreia Oliveira, Julia Richter, Shunichi Takeda, Shunsuke Kobayashi, Masataka Tsuda, Mariela Artola-Borán, Joanna I. Loizou, Xueqing Zou, and University of Zurich
- Subjects
Exonuclease ,Methylnitronitrosoguanidine ,congenital, hereditary, and neonatal diseases and abnormalities ,exonuclease ,DNA repair ,610 Medicine & health ,EXO1 ,DNA Mismatch Repair ,Cell Line ,Avian Proteins ,03 medical and health sciences ,Exonuclease 1 ,0302 clinical medicine ,Germline mutation ,FAN1 ,PMS2 ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Endodeoxyribonucleases ,Thionucleosides ,biology ,Base Sequence ,Guanosine ,10061 Institute of Molecular Cancer Research ,MLH1 ,Cell Biology ,mutational signature ,MSH6 ,Multifunctional Enzymes ,digestive system diseases ,Cell biology ,mismatch repair ,Exodeoxyribonucleases ,HEK293 Cells ,MSH2 ,030220 oncology & carcinogenesis ,Mutation ,biology.protein ,570 Life sciences ,DNA mismatch repair ,Chickens ,Research Article - Abstract
Germline mutations in the mismatch repair (MMR) genes MSH2, MSH6, MLH1, and PMS2 are linked to cancer of the colon and other organs, characterized by microsatellite instability and a large increase in mutation frequency. Unexpectedly, mutations in EXO1, encoding the only exonuclease genetically implicated in MMR, are not linked to familial cancer and cause a substantially weaker mutator phenotype. This difference could be explained if eukaryotic cells possessed additional exonucleases redundant with EXO1. Analysis of the MLH1 interactome identified FANCD2-associated nuclease 1 (FAN1), a novel enzyme with biochemical properties resembling EXO1. We now show that FAN1 efficiently substitutes for EXO1 in MMR assays and that this functional complementation is modulated by its interaction with MLH1. FAN1 also contributes to MMR in vivo; cells lacking both EXO1 and FAN1 have an MMR defect and display resistance to N-methyl-N-nitrosourea (MNU) and 6-thioguanine (TG). Moreover, FAN1 loss amplifies the mutational profile of EXO1-deficient cells, suggesting that the two nucleases act redundantly in the same antimutagenic pathway. However, the increased drug resistance and mutator phenotype of FAN1/EXO1-deficient cells are less prominent than those seen in cells lacking MSH6 or MLH1. Eukaryotic cells thus apparently possess additional mechanisms that compensate for the loss of EXO1., Cellular and Molecular Biology, 41 (9), ISSN:0145-5680, ISSN:1165-158X
- Published
- 2021
29. The role of the changing human microbiome in the asthma pandemic
- Author
-
Xiaozhou Zhang, Timothy C. Borbet, Martin J. Blaser, Anne Müller, University of Zurich, and Müller, Anne
- Subjects
0301 basic medicine ,Allergy ,Immunology ,610 Medicine & health ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Risk Factors ,Pandemic ,Humans ,Immunology and Allergy ,Medicine ,Microbiome ,Pandemics ,Asthma ,2403 Immunology ,Toll-like receptor ,Helicobacter pylori ,business.industry ,Microbiota ,10061 Institute of Molecular Cancer Research ,Human microbiome ,Allergens ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,2723 Immunology and Allergy ,570 Life sciences ,biology ,Disease Susceptibility ,business - Abstract
Asthma and allergy incidence continue to increase globally. We have made significant strides in treating disease, but it is becoming more apparent that we need to advance our knowledge into the origins of asthmatic disease. Much recent work has indicated that microbiome composition influences immune regulation and that multiple health care factors have driven a loss in microbiome diversity in modern human populations. Evidence is growing of microbiota-driven influences on immune development, asthma susceptibility, and asthma pathogenesis. The focus of this review is to highlight the strides the field has made in characterizing the constituents of the human gastrointestinal microbiota, such as Helicobacter pylori, other members of the neonatal intestinal microbiota, and microbial peptides and metabolites that influence host immunity and immune response to allergens. As we delve further into this field of research, the goal will be to find actionable and clinical interventions to identify at-risk populations earlier to prevent disease onset. Manipulation of the host microbial community during infancy might be an especially promising approach.
- Published
- 2019
- Full Text
- View/download PDF
30. T4SS-dependent TLR5 activation by Helicobacter pylori infection
- Author
-
Christina Falkeis-Veits, Michael Vieth, Steffen Backert, Anne Müller, Matthias Neddermann, Sujay Chattopadhyay, Nicole Tegtmeyer, Mark Brönstrup, Isabelle C. Arnold, Suneesh Kumar Pachathundikandi, Minsun Hong, Judith Lind, Heinrich Sticht, Werner Tegge, University of Zurich, Backert, Steffen, and HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany.
- Subjects
0301 basic medicine ,Biopsy ,General Physics and Astronomy ,medicine.disease_cause ,Mice ,0302 clinical medicine ,lcsh:Science ,Receptor ,Mice, Knockout ,Multidisciplinary ,10061 Institute of Molecular Cancer Research ,NF-kappa B ,3100 General Physics and Astronomy ,3. Good health ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,Female ,Pathogens ,Signal Transduction ,Science ,Virulence ,610 Medicine & health ,1600 General Chemistry ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Microbiology ,Helicobacter Infections ,Type IV Secretion Systems ,03 medical and health sciences ,Immune system ,Bacterial Proteins ,1300 General Biochemistry, Genetics and Molecular Biology ,medicine ,Animals ,Humans ,Secretion ,Helicobacter pylori ,Activator (genetics) ,Pathogenic bacteria ,General Chemistry ,biology.organism_classification ,Toll-like receptors ,Disease Models, Animal ,Toll-Like Receptor 5 ,030104 developmental biology ,TLR5 ,Gastric Mucosa ,570 Life sciences ,biology ,lcsh:Q - Abstract
Toll-like receptor TLR5 recognizes a conserved domain, termed D1, that is present in flagellins of several pathogenic bacteria but not in Helicobacter pylori. Highly virulent H. pylori strains possess a type IV secretion system (T4SS) for delivery of virulence factors into gastric epithelial cells. Here, we show that one of the H. pylori T4SS components, protein CagL, can act as a flagellin-independent TLR5 activator. CagL contains a D1-like motif that mediates adherence to TLR5+ epithelial cells, TLR5 activation, and downstream signaling in vitro. TLR5 expression is associated with H. pylori infection and gastric lesions in human biopsies. Using Tlr5-knockout and wild-type mice, we show that TLR5 is important for efficient control of H. pylori infection. Our results indicate that CagL, by activating TLR5, may modulate immune responses to H. pylori., Toll-like receptor TLR5 recognizes a domain, D1, that is present in flagellins of several pathogenic bacteria but not in Helicobacter pylori. Here, the authors show that TLR5 can be activated independently of flagellin by a component of the H. pylori type IV secretion system that contains a D1-like motif.
- Published
- 2019
31. Helicobacter pylori-controlled c-Abl localization promotes cell migration and limits apoptosis
- Author
-
Posselt, Gernot, Wiesauer, Maria, Chichirau, Bianca E, Engler, Daniela, Krisch, Linda M, Gadermaier, Gabriele, Briza, Peter, Schneider, Sabine, Boccellato, Francesco, Meyer, Thomas F, Hauser-Kronberger, Cornelia, Neureiter, Daniel, Müller, Anne, Wessler, Silja, University of Zurich, and Wessler, Silja
- Subjects
1303 Biochemistry ,lcsh:Medicine ,610 Medicine & health ,Apoptosis ,Models, Biological ,Helicobacter Infections ,1307 Cell Biology ,Cell Movement ,Cell Line, Tumor ,hemic and lymphatic diseases ,1312 Molecular Biology ,Humans ,Phosphorylation ,PKC ,lcsh:QH573-671 ,Phosphotyrosine ,Proto-Oncogene Proteins c-abl ,Protein Kinase C ,Cancer ,Helicobacter pylori ,lcsh:Cytology ,Research ,10061 Institute of Molecular Cancer Research ,lcsh:R ,Motility ,C-Abl ,βHBP ,Protein Transport ,Phosphothreonine ,Gastritis ,570 Life sciences ,biology - Abstract
Background Deregulated c-Abl activity has been intensively studied in a variety of solid tumors and leukemia. The class-I carcinogen Helicobacter pylori (Hp) activates the non-receptor tyrosine kinase c-Abl to phosphorylate the oncoprotein cytotoxin-associated gene A (CagA). The role of c-Abl in CagA-dependent pathways is well established; however, the knowledge of CagA-independent c-Abl processes is scarce. Methods c-Abl phosphorylation and localization were analyzed by immunostaining and immunofluorescence. Interaction partners were identified by tandem-affinity purification. Cell elongation and migration were analyzed in transwell-filter experiments. Apoptosis and cell survival were examined by FACS analyses and MTT assays. In mice experiments and human biopsies, the involvement of c-Abl in Hp pathogenesis was investigated. Results Here, we investigated the activity and subcellular localization of c-Abl in vitro and in vivo and unraveled the contribution of c-Abl in CagA-dependent and -independent pathways to gastric Hp pathogenesis. We report a novel mechanism and identified strong c-Abl threonine 735 phosphorylation (pAblT735) mediated by the type-IV secretion system (T4SS) effector D-glycero-β-D-manno-heptose-1,7-bisphosphate (βHBP) and protein kinase C (PKC) as a new c-Abl kinase. pAblT735 interacted with 14–3-3 proteins, which caused cytoplasmic retention of c-Abl, where it potentiated Hp-mediated cell elongation and migration. Further, the nuclear exclusion of pAblT735 attenuated caspase-8 and caspase-9-dependent apoptosis. Importantly, in human patients suffering from Hp-mediated gastritis c-Abl expression and pAblT735 phosphorylation were drastically enhanced as compared to type C gastritis patients or healthy individuals. Pharmacological inhibition using the selective c-Abl kinase inhibitor Gleevec confirmed that c-Abl plays an important role in Hp pathogenesis in a murine in vivo model. Conclusions In this study, we identified a novel regulatory mechanism in Hp-infected gastric epithelial cells by which Hp determines the subcellular localization of activated c-Abl to control Hp-mediated EMT-like processes while decreasing cell death. Electronic supplementary material The online version of this article (10.1186/s12964-019-0323-9) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
32. PrimPol‐mediated repriming facilitates replication traverse of DNA interstrand crosslinks
- Author
-
Susana Llanos, Karun Mutreja, Patricia Ubieto-Capella, Samuel Miguez, Daniel González-Acosta, Juan Méndez, Fernando García, Elena Blanco-Romero, Javier Munoz, Luis Blanco, Massimo Lopes, University of Zurich, Méndez, Juan, Ministerio de Ciencia, Innovación y Universidades (España), and Swiss National Science Foundation
- Subjects
Male ,DNA Repair ,DNA-Directed DNA Polymerase ,Interstrand crosslink ,TOP3A ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Transcription (biology) ,Fanconi anemia ,2400 General Immunology and Microbiology ,DNA Crosslinking ,Translocase ,FANCM ,Mammals ,0303 health sciences ,biology ,General Neuroscience ,10061 Institute of Molecular Cancer Research ,2800 General Neuroscience ,Articles ,Cell biology ,Female ,Primase ,RPA ,DNA Replication ,ICL repair ,610 Medicine & health ,DNA Primase ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,1300 General Biochemistry, Genetics and Molecular Biology ,1312 Molecular Biology ,medicine ,Animals ,Humans ,ICL traverse ,Molecular Biology ,030304 developmental biology ,General Immunology and Microbiology ,DNA Helicases ,DNA Replication, Repair & Recombination ,DNA ,medicine.disease ,Multifunctional Enzymes ,chemistry ,biology.protein ,570 Life sciences ,PrimPol ,030217 neurology & neurosurgery - Abstract
DNA interstrand crosslinks (ICLs) induced by endogenous aldehydes or chemotherapeutic agents interfere with essential processes such as replication and transcription. ICL recognition and repair by the Fanconi Anemia pathway require the formation of an X‐shaped DNA structure that may arise from convergence of two replication forks at the crosslink or traversing of the lesion by a single replication fork. Here, we report that ICL traverse strictly requires DNA repriming events downstream of the lesion, which are carried out by PrimPol, the second primase‐polymerase identified in mammalian cells after Polα/Primase. The recruitment of PrimPol to the vicinity of ICLs depends on its interaction with RPA, but not on FANCM translocase or the BLM/TOP3A/RMI1‐2 (BTR) complex that also participate in ICL traverse. Genetic ablation of PRIMPOL makes cells more dependent on the fork convergence mechanism to initiate ICL repair, and PRIMPOL KO cells and mice display hypersensitivity to ICL‐inducing drugs. These results open the possibility of targeting PrimPol activity to enhance the efficacy of chemotherapy based on DNA crosslinking agents., A specialized human primase involved in DNA damage tolerance facilitates continuous DNA synthesis past ICL lesions, and their post‐replicative repair via the Fanconi Anemia pathway.
- Published
- 2021
- Full Text
- View/download PDF
33. The metabolome and lipidome of colorectal adenomas and cancers
- Author
-
Laczko, Endre, Manser, Christine, Marra, Giancarlo, and University of Zurich
- Subjects
10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health - Published
- 2021
- Full Text
- View/download PDF
34. ATG5 promotes eosinopoiesis but inhibits eosinophil effector functions
- Author
-
Anne Müller, Nina Germic, Anne Angelillo-Scherrer, Sara Calzavarini, Hans-Uwe Simon, Carsten Riether, Meike Claus, Isabelle C. Arnold, Kevin Oberson, Charaf Benarafa, Aref Hosseini, Darko Stojkov, Shida Yousefi, University of Zurich, and Simon, Hans-Uwe
- Subjects
Receptor, Platelet-Derived Growth Factor alpha ,1303 Biochemistry ,Oncogene Proteins, Fusion ,2720 Hematology ,Biochemistry ,Cell Degranulation ,Autophagy-Related Protein 5 ,1307 Cell Biology ,Phagocytes, Granulocytes, and Myelopoiesis ,Leukocyte Count ,Mice ,Bone Marrow ,Hypereosinophilic Syndrome ,Eosinophilia ,610 Medicine & health ,Cells, Cultured ,Mice, Knockout ,Myelopoiesis ,630 Agriculture ,10061 Institute of Molecular Cancer Research ,Degranulation ,Enterobacteriaceae Infections ,Hematology ,respiratory system ,medicine.anatomical_structure ,medicine.symptom ,MAP Kinase Signaling System ,Immunology ,Mice, Transgenic ,Biology ,Colony-Forming Units Assay ,Immune system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Interleukin 5 ,mRNA Cleavage and Polyadenylation Factors ,Chronic eosinophilic leukemia ,Eosinophil differentiation ,2403 Immunology ,Cell Biology ,Eosinophil ,medicine.disease ,Eosinophils ,Citrobacter rodentium ,570 Life sciences ,biology ,Bone marrow ,CRISPR-Cas Systems ,Interleukin-5 - Abstract
Eosinophils are white blood cells contributing to the regulation of immunity and they are involved in the pathogenesis of numerous inflammatory diseases. In contrast to other cells of the immune system, no information is available about the role of autophagy in eosinophil differentiation and functions. In order to study the autophagic pathway in eosinophils, we generated conditional knockout mice in which Atg5 is deleted within the eosinophil lineage only (designated Atg5eoΔ mice). Eosinophilia was provoked by crossbreeding Atg5eoΔ mice with Il5 (IL-5) overexpressing transgenic mice (designated Atg5eoΔIl5tg mice). Deletion of Atg5 in eosinophils resulted in a dramatic reduction in the number of mature eosinophils in blood and in an increase of immature eosinophils in the bone marrow. Atg5-knockout eosinophil precursors exhibited reduced proliferation under both in vitro and in vivo conditions, but no increased cell death. Moreover, reduced differentiation of eosinophils in the absence of Atg5 was also seen in mouse and human models of chronic eosinophilic leukemia. Atg5-knockout blood eosinophils demonstrated augmented levels of degranulation and bacterial killing in vitro. Moreover, in an experimental in vivo model, we observed that Atg5eoΔ mice achieve better clearance of the local and systemic bacterial infection with Citrobacter (C.) rodentium. Evidence for increased degranulation of ATG5low-expressing human eosinophils was also obtained in both tissues and blood. Taken together, mouse and human eosinophil hematopoiesis and effector functions are regulated by ATG5 which controls the amplitude of overall antibacterial eosinophil immune responses.
- Published
- 2021
- Full Text
- View/download PDF
35. Stereo- and regiodefined DNA-encoded chemical libraries enable efficient tumour-targeting applications
- Author
-
Dario Neri, Renier Myburgh, Alessandro A. Sartori, Roberto De Luca, Mustafa Moroglu, Richard A. Lerner, Jörg Scheuermann, Peter K. Vogt, Christian Pellegrino, Anika Trenner, Nicholas Favalli, Nour L. Mozaffari, Jacopo Millul, Markus G. Manz, Stuart J. Conway, Su Yang, Gabriele Bassi, Samuele Cazzamalli, and University of Zurich
- Subjects
T-Lymphocytes ,General Chemical Engineering ,Immunoglobulin Variable Region ,Tumour targeting ,610 Medicine & health ,Computational biology ,010402 general chemistry ,01 natural sciences ,Fluorescence ,Article ,Mice ,chemistry.chemical_compound ,Drug Delivery Systems ,Antigen ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Animals ,Humans ,Gene Library ,chemistry.chemical_classification ,Receptors, Chimeric Antigen ,010405 organic chemistry ,Ligand ,Chemistry ,10061 Institute of Molecular Cancer Research ,Regioselectivity ,DNA ,Neoplasms, Experimental ,General Chemistry ,Chimeric antigen receptor ,0104 chemical sciences ,3. Good health ,Enzyme ,Microscopy, Fluorescence ,10032 Clinic for Oncology and Hematology ,570 Life sciences ,biology - Abstract
The encoding of chemical compounds with amplifiable DNA tags facilitates the discovery of small-molecule ligands for proteins. To investigate the impact of stereo- and regiochemistry on ligand discovery, we synthesized a DNA-encoded library of 670,752 derivatives based on 2-azido-3-iodophenylpropionic acids. The library was selected against multiple proteins and yielded specific ligands. The selection fingerprints obtained for a set of protein targets of pharmaceutical relevance clearly showed the preferential enrichment of ortho-, meta- or para-regioisomers, which was experimentally verified by affinity measurements in the absence of DNA. The discovered ligands included novel selective enzyme inhibitors and binders to tumour-associated antigens, which enabled conditional chimeric antigen receptor T-cell activation and tumour targeting. [Figure not available: see fulltext.] ISSN:1755-4349 ISSN:1755-4330
- Published
- 2021
- Full Text
- View/download PDF
36. Human CtIP: A 'double agent' in DNA repair and tumorigenesis
- Author
-
Alessandro A. Sartori, Nour L. Mozaffari, Fabio Pagliarulo, University of Zurich, and Sartori, Alessandro A
- Subjects
0301 basic medicine ,Genome instability ,DNA Repair ,DNA repair ,Carcinogenesis ,610 Medicine & health ,Biology ,medicine.disease_cause ,Replication fork protection ,1309 Developmental Biology ,1307 Cell Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,10061 Institute of Molecular Cancer Research ,Retinoblastoma protein ,Cell Biology ,DNA Repair Pathway ,G2-M DNA damage checkpoint ,3. Good health ,030104 developmental biology ,biology.protein ,Cancer research ,570 Life sciences ,biology ,Homologous recombination ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Human CtIP was originally identified as an interactor of the retinoblastoma protein and BRCA1, two bona fide tumour suppressors frequently mutated in cancer. CtIP is renowned for its role in the resection of DNA double-strand breaks (DSBs) during homologous recombination, a largely error-free DNA repair pathway crucial in maintaining genome integrity. However, CtIP-dependent DNA end resection is equally accountable for alternative end-joining, a mutagenic DSB repair mechanism implicated in oncogenic chromosomal translocations. In addition, CtIP contributes to transcriptional regulation of G1/S transition, DNA damage checkpoint signalling, and replication fork protection pathways. In this review, we present a perspective on the current state of knowledge regarding the tumour-suppressive and oncogenic properties of CtIP and provide an overview of their relevance for cancer development, progression, and therapy.
- Published
- 2021
37. RAD51 Inhibition Induces R-Loop Formation in Early G1 Phase of the Cell Cycle
- Author
-
Zuzana Nascakova, Vaclav Urban, Barbora Boleslavska, Edita Vlachova, Jana Dobrovolna, Anna Oravetzova, Pavel Janscak, University of Zurich, Janscak, Pavel, and Dobrovolna, Jana
- Subjects
1503 Catalysis ,RAD51 ,Origin Recognition Complex ,1607 Spectroscopy ,610 Medicine & health ,B02 inhibitor ,Pre-replication complex ,Article ,Catalysis ,Immunoglobulin Class Switch Recombination ,Inorganic Chemistry ,lcsh:Chemistry ,chemistry.chemical_compound ,Cell Line, Tumor ,Chromosomal Instability ,1312 Molecular Biology ,1706 Computer Science Applications ,Humans ,Physical and Theoretical Chemistry ,Enzyme Inhibitors ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,DNA synthesis ,Chemistry ,1604 Inorganic Chemistry ,Organic Chemistry ,10061 Institute of Molecular Cancer Research ,G1 Phase ,General Medicine ,DNA ,G1 phase of the cell cycle ,Cell cycle ,Computer Science Applications ,Cell biology ,origin of replication ,pre-replication complex ,lcsh:Biology (General) ,lcsh:QD1-999 ,570 Life sciences ,biology ,R-loop ,Rad51 Recombinase ,R-Loop Structures ,Homologous recombination ,1606 Physical and Theoretical Chemistry ,Mitochondrial DNA replication ,1605 Organic Chemistry - Abstract
R-loops are three-stranded structures generated by annealing of nascent transcripts to the template DNA strand, leaving the non-template DNA strand exposed as a single-stranded loop. Although R-loops play important roles in physiological processes such as regulation of gene expression, mitochondrial DNA replication, or immunoglobulin class switch recombination, dysregulation of the R-loop metabolism poses a threat to the stability of the genome. A previous study in yeast has shown that the homologous recombination machinery contributes to the formation of R-loops and associated chromosome instability. On the contrary, here, we demonstrate that depletion of the key homologous recombination factor, RAD51, as well as RAD51 inhibition by the B02 inhibitor did not prevent R-loop formation induced by the inhibition of spliceosome assembly in human cells. However, we noticed that treatment of cells with B02 resulted in RAD51-dependent accumulation of R-loops in an early G1 phase of the cell cycle accompanied by a decrease in the levels of chromatin-bound ORC2 protein, a component of the pre-replication complex, and an increase in DNA synthesis. Our results suggest that B02-induced R-loops might cause a premature origin firing.
- Published
- 2021
38. An Antibiotic-Impacted Microbiota Compromises the Development of Colonic Regulatory T Cells and Predisposes to Dysregulated Immune Responses
- Author
-
Zhang, Xiaozhou, Borbet, Timothy C, Fallegger, Angela, Wipperman, Matthew F, Blaser, Martin J, Müller, Anne, University of Zurich, and Blaser, Martin J
- Subjects
10061 Institute of Molecular Cancer Research ,2404 Microbiology ,2406 Virology ,570 Life sciences ,biology ,610 Medicine & health ,Microbiology ,QR1-502 - Abstract
The assembly of microbial communities that populate all mucosal surfaces of the human body begins right after birth. This process is prone to disruption as newborns and young infants are increasingly exposed to antibiotics, both deliberately for therapeutic purposes, and as a consequence of transmaternal exposure.
- Published
- 2021
- Full Text
- View/download PDF
39. MDM2 binds and ubiquitinates PARP1 to enhance DNA replication fork progression
- Author
-
Giansanti, Celeste, Manzini, Valentina, Dickmanns, Antje, Dickmanns, Achim, Palumbieri, Maria Dilia, Sanchi, Andrea, Kienle, Simon Maria, Rieth, Sonja, Scheffner, Martin, Lopes, Massimo, Dobbelstein, Matthias, University of Zurich, and Dobbelstein, Matthias
- Subjects
DNA Replication ,1300 General Biochemistry, Genetics and Molecular Biology ,ddc:570 ,MDM2, PARP1, ubiquitin, DNA replication, replication fork reversal, RECQ1, PRIMPOL, p53, poly ADP ribose ,10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health ,DNA ,DNA Primase ,Tumor Suppressor Protein p53 ,neoplasms ,General Biochemistry, Genetics and Molecular Biology ,DNA Damage - Abstract
The MDM2 oncoprotein antagonizes the tumor suppressor p53 by physical interaction and ubiquitination. However, it also sustains the progression of DNA replication forks, even in the absence of functional p53. Here, we show that MDM2 binds, inhibits, ubiquitinates, and destabilizes poly(ADP-ribose) polymerase 1 (PARP1). When cellular MDM2 levels are increased, this leads to accelerated progression of DNA replication forks, much like pharmacological inhibition of PARP1. Conversely, overexpressed PARP1 restores normal fork progression despite elevated MDM2. Strikingly, MDM2 profoundly reduces the frequency of fork reversal, revealed as four-way junctions through electron microscopy. Depletion of RECQ1 or the primase/polymerase (PRIMPOL) reverses the MDM2-mediated acceleration of the nascent DNA elongation rate. MDM2 also increases the occurrence of micronuclei, and it exacerbates camptothecin-induced cell death. In conclusion, high MDM2 levels phenocopy PARP inhibition in modulation of fork restart, representing a potential vulnerability of cancer cells. published
- Published
- 2022
- Full Text
- View/download PDF
40. The GM-CSF–IRF5 signaling axis in eosinophils promotes antitumor immunity through activation of type 1 T cell responses
- Author
-
Michael Bauer, Mariela Artola-Borán, Manfred Kopf, Alexandar Tzankov, Katrin Bertram, Anne Müller, Ziva Frangez, Hans-Uwe Simon, Shida Yousefi, Alessandra Gurtner, Isabelle C. Arnold, Burkhard Becher, University of Zurich, and Arnold, Isabelle C
- Subjects
Male ,Transcription, Genetic ,Carcinogenesis ,Lymphocyte Activation ,0302 clinical medicine ,Neoplasms ,Tumor Microenvironment ,Immunology and Allergy ,Cytotoxic T cell ,Transgenes ,Immune Checkpoint Inhibitors ,0303 health sciences ,10061 Institute of Molecular Cancer Research ,Mucosal Immunology ,3. Good health ,Interleukin-10 ,Intestines ,Interleukin 10 ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Interferon Regulatory Factors ,2723 Immunology and Allergy ,Female ,Signal Transduction ,Tumor Immunology ,Adenoma ,STAT3 Transcription Factor ,Cell type ,T cell ,Immunology ,610 Medicine & health ,Biology ,Article ,03 medical and health sciences ,Immunity ,Cell Line, Tumor ,Genetic model ,medicine ,Animals ,Humans ,030304 developmental biology ,Cell Proliferation ,2403 Immunology ,Granulocyte-Macrophage Colony-Stimulating Factor ,Eosinophil ,Th1 Cells ,Survival Analysis ,Eosinophils ,Mice, Inbred C57BL ,Cancer research ,570 Life sciences ,biology ,Lymph Nodes ,Interleukin-5 ,CD8 - Abstract
Arnold et al. report that eosinophils in intestinal tumors are conditioned by GM-CSF to promote antitumor immunity through the activation of Th1 and CD8+ T cell responses. GM-CSF activates IRF5 and can be administered recombinantly to reduce tumor growth. Colorectal cancer patients exhibiting high intratumoral eosinophil infiltration also have better prognosis., The depletion of eosinophils represents an efficient strategy to alleviate allergic asthma, but the consequences of prolonged eosinophil deficiency for human health remain poorly understood. We show here that the ablation of eosinophils severely compromises antitumor immunity in syngeneic and genetic models of colorectal cancer (CRC), which can be attributed to defective Th1 and CD8+ T cell responses. The specific loss of GM-CSF signaling or IRF5 expression in the eosinophil compartment phenocopies the loss of the entire lineage. GM-CSF activates IRF5 in vitro and in vivo and can be administered recombinantly to improve tumor immunity. IL-10 counterregulates IRF5 activation by GM-CSF. CRC patients whose tumors are infiltrated by large numbers of eosinophils also exhibit robust CD8 T cell infiltrates and have a better prognosis than patients with eosinophillow tumors. The combined results demonstrate a critical role of eosinophils in tumor control in CRC and introduce the GM-CSF–IRF5 axis as a critical driver of the antitumor activities of this versatile cell type.
- Published
- 2020
- Full Text
- View/download PDF
41. Ubiquitin phosphorylation at Thr12 modulates the DNA damage response
- Author
-
Dario Neri, Huib Ovaa, Sibylle Burger, Tatiana Gubser, Matthias Altmeyer, Marco Gatti, Alessandra Villa, Benoît Bragantini, Monique P. C. Mulder, Lorenza Penengo, Maria Victoria Botuyan, Franziska Walser, Georges Mer, and University of Zurich
- Subjects
Threonine ,DNA End-Joining Repair ,DNA Repair ,DNA repair ,DNA damage ,Ubiquitin-Protein Ligases ,RAD51 ,610 Medicine & health ,Article ,Cell Line ,Deubiquitinating enzyme ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Cell Line, Tumor ,Histone H2A ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Homologous Recombination ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,10061 Institute of Molecular Cancer Research ,Intracellular Signaling Peptides and Proteins ,Ubiquitination ,Nuclear Proteins ,DNA ,Cell Biology ,10226 Department of Molecular Mechanisms of Disease ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Histone ,biology.protein ,570 Life sciences ,Tumor Suppressor p53-Binding Protein 1 ,030217 neurology & neurosurgery ,DNA Damage ,Signal Transduction - Abstract
The ubiquitin system regulates the DNA damage response (DDR) by modifying histone H2A at Lys15 (H2AK15ub) and triggering downstream signaling events. Here, we find that phosphorylation of ubiquitin at Thr12 (pUbT12) controls the DDR by inhibiting the function of 53BP1, a key factor for DNA double-strand break repair by non-homologous end joining (NHEJ). Detectable as a chromatin modification on H2AK15ub, pUbT12 accumulates in nuclear foci and is increased upon DNA damage. Mutating Thr12 prevents the removal of ubiquitin from H2AK15ub by USP51 deubiquitinating enzyme, leading to a pronounced accumulation of ubiquitinated chromatin. Chromatin modified by pUbT12 is inaccessible to 53BP1 but permissive to the homologous recombination (HR) proteins RNF169, RAD51, and the BRCA1/BARD1 complex. Phosphorylation of ubiquitin at Thr12 in the chromatin context is a new histone mark, H2AK15pUbT12, that regulates the DDR by hampering the activity of 53BP1 at damaged chromosomes.
- Published
- 2020
- Full Text
- View/download PDF
42. A Single-Stranded DNA-Encoded Chemical Library Based on a Stereoisomeric Scaffold Enables Ligand Discovery by Modular Assembly of Building Blocks
- Author
-
Bassi, Gabriele, Favalli, Nicholas, Vuk, Miriam, Catalano, Marco, Martinelli, Adriano, Trenner, Anika, Porro, Antonio, Yang, Su, Tham, Chuin Lean, Moroglu, Mustafa, Yue, Wyatt W, Conway, Stuart J, Vogt, Peter K, Sartori, Alessandro A, Scheuermann, Jörg, Neri, Dario, University of Zurich, Scheuermann, Jörg, and Neri, Dario
- Subjects
10061 Institute of Molecular Cancer Research ,2200 General Engineering ,570 Life sciences ,biology ,610 Medicine & health ,2701 Medicine (miscellaneous) ,1500 General Chemical Engineering ,1301 Biochemistry, Genetics and Molecular Biology (miscellaneous) ,2500 General Materials Science ,3100 General Physics and Astronomy - Published
- 2020
- Full Text
- View/download PDF
43. Functional radiogenetic profiling implicates ERCC6L2 in non-homologous end joining
- Author
-
Francica, Paola, Mutlu, Merve, Blomen, Vincent A, Oliveira, Catarina, Nowicka, Zuzanna, Trenner, Anika, Gerhards, Nora M, Bouwman, Peter, Stickel, Elmer, Hekkelman, Maarten L, Lingg, Lea, Klebic, Ismar, van de Ven, Marieke, de Korte-Grimmerink, Renske, Howald, Denise, Jonkers, Jos, Sartori, Alessandro A, Fendler, Wojciech, Chapman, J Ross, Brummelkamp, Thijn, Rottenberg, Sven, University of Zurich, and Rottenberg, Sven
- Subjects
Mice ,DNA End-Joining Repair ,lcsh:Biology (General) ,630 Agriculture ,1300 General Biochemistry, Genetics and Molecular Biology ,10061 Institute of Molecular Cancer Research ,DNA Helicases ,Animals ,Humans ,570 Life sciences ,biology ,610 Medicine & health ,lcsh:QH301-705.5 - Abstract
Summary: Using genome-wide radiogenetic profiling, we functionally dissect vulnerabilities of cancer cells to ionizing radiation (IR). We identify ERCC6L2 as a major determinant of IR response, together with classical DNA damage response genes and members of the recently identified shieldin and CTC1-STN1-TEN1 (CST) complexes. We show that ERCC6L2 contributes to non-homologous end joining (NHEJ), and it may exert this function through interactions with SFPQ. In addition to causing radiosensitivity, ERCC6L2 loss restores DNA end resection and partially rescues homologous recombination (HR) in BRCA1-deficient cells. As a consequence, ERCC6L2 deficiency confers resistance to poly (ADP-ribose) polymerase (PARP) inhibition in tumors deficient for both BRCA1 and p53. Moreover, we show that ERCC6L2 mutations are found in human tumors and correlate with a better overall survival in patients treated with radiotherapy (RT); this finding suggests that ERCC6L2 is a predictive biomarker of RT response.
- Published
- 2020
- Full Text
- View/download PDF
44. An 'expressionistic' look at serrated precancerous colorectal lesions
- Author
-
Marra, Giancarlo, University of Zurich, and Marra, Giancarlo
- Subjects
Adenoma ,Male ,Pathology ,medicine.medical_specialty ,Histology ,Colorectal cancer ,Colon ,Rectum ,610 Medicine & health ,In situ hybridization ,Biology ,2722 Histology ,Adenomatous polyp ,Pathology and Forensic Medicine ,Lesion ,Adenomatous Polyps ,Gene expression ,lcsh:Pathology ,medicine ,Humans ,RNA, Messenger ,Traditional serrated adenoma ,Hyperplastic polyp ,Tissue staining markers ,Research ,10061 Institute of Molecular Cancer Research ,Sessile serrated lesion ,RNA ,General Medicine ,medicine.disease ,2734 Pathology and Forensic Medicine ,medicine.anatomical_structure ,HOXD13 ,Hyperplastic Polyp ,570 Life sciences ,biology ,Female ,medicine.symptom ,Colorectal Neoplasms ,Precancerous Conditions ,Biomarkers ,lcsh:RB1-214 - Abstract
Background Approximately 60% of colorectal cancer (CRC) precursor lesions are the genuinely-dysplastic conventional adenomas (cADNs). The others include hyperplastic polyps (HPs), sessile serrated lesions (SSL), and traditional serrated adenomas (TSAs), subtypes of a class of lesions collectively referred to as “serrated.” Endoscopic and histologic differentiation between cADNs and serrated lesions, and between serrated lesion subtypes can be difficult. Methods We used in situ hybridization to verify the expression patterns in CRC precursors of 21 RNA molecules that appear to be promising differentiation markers on the basis of previous RNA sequencing studies. Results SSLs could be clearly differentiated from cADNs by the expression patterns of 9 of the 12 RNAs tested for this purpose (VSIG1, ANXA10, ACHE, SEMG1, AQP5, LINC00520, ZIC5/2, FOXD1, NKD1). Expression patterns of all 9 in HPs were similar to those in SSLs. Nine putatively HP-specific RNAs were also investigated, but none could be confirmed as such: most (e.g., HOXD13 and HOXB13), proved instead to be markers of the normal mucosa in the distal colon and rectum, where most HPs arise. TSAs displayed mixed staining patterns reflecting the presence of serrated and dysplastic glands in the same lesion. Conclusions Using a robust in situ hybridization protocol, we identified promising tissue-staining markers that, if validated in larger series of lesions, could facilitate more precise histologic classification of CRC precursors and, consequently, more tailored clinical follow-up of their carriers. Our findings should also fuel functional studies on the pathogenic significance of specific gene expression alterations in the initiation and evolution of CRC precursor subtypes.
- Published
- 2020
45. Interferon-stimulated gene 15 accelerates replication fork progression inducing chromosomal breakage
- Author
-
Franziska Walser, Klaus-Peter Knobeloch, Lorenza Penengo, Nikola Djoric, Maria Chiara Raso, Fabian Schmid, Sandra Hess, Sibylle Burger, University of Zurich, and Penengo, Lorenza
- Subjects
DNA Replication ,Time Factors ,DNA damage ,Antineoplastic Agents ,Bone Neoplasms ,610 Medicine & health ,Antiviral Agents ,1307 Cell Biology ,03 medical and health sciences ,chemistry.chemical_compound ,DNA biology ,0302 clinical medicine ,Proliferating Cell Nuclear Antigen ,Report ,Humans ,Spotlight ,Gene ,Ubiquitins ,030304 developmental biology ,Cancer ,Osteosarcoma ,0303 health sciences ,Dose-Response Relationship, Drug ,RecQ Helicases ,biology ,DNA synthesis ,10061 Institute of Molecular Cancer Research ,DNA replication ,Helicase ,Chromosome Breakage ,DNA, Neoplasm ,Cell Biology ,DNA Replication Fork ,Immunity, Innate ,3. Good health ,Proliferating cell nuclear antigen ,Cell biology ,HEK293 Cells ,chemistry ,030220 oncology & carcinogenesis ,Interferon Type I ,MCF-7 Cells ,biology.protein ,Cytokines ,570 Life sciences ,DNA ,DNA Damage ,HeLa Cells - Abstract
Raso et al. find that high levels of interferon-stimulated gene 15 (ISG15), which is very frequent in cancer and robustly induced by pathogen infection, accelerate DNA replication fork progression, impacting genome stability and response to chemotherapy., DNA replication is highly regulated by the ubiquitin system, which plays key roles upon stress. The ubiquitin-like modifier ISG15 (interferon-stimulated gene 15) is induced by interferons, bacterial and viral infection, and DNA damage, but it is also constitutively expressed in many types of cancer, although its role in tumorigenesis is still largely elusive. Here, we show that ISG15 localizes at the replication forks, in complex with PCNA and the nascent DNA, where it regulates DNA synthesis. Indeed, high levels of ISG15, intrinsic or induced by interferon-β, accelerate DNA replication fork progression, resulting in extensive DNA damage and chromosomal aberrations. This effect is largely independent of ISG15 conjugation and relies on ISG15 functional interaction with the DNA helicase RECQ1, which promotes restart of stalled replication forks. Additionally, elevated ISG15 levels sensitize cells to cancer chemotherapeutic treatments. We propose that ISG15 up-regulation exposes cells to replication stress, impacting genome stability and response to genotoxic drugs.
- Published
- 2020
- Full Text
- View/download PDF
46. The iron-sulphur cluster in human DNA2 is required for all biochemical activities of DNA2
- Author
-
Mariotti, Laura, Wild, Sebastian, Brunoldi, Giulia, Piceni, Alessandra, Ceppi, Ilaria, Kummer, Sandra, Lutz, Richard E, Cejka, Petr, Gari, Kerstin, University of Zurich, Mariotti, Laura, and Gari, Kerstin
- Subjects
1300 General Biochemistry, Genetics and Molecular Biology ,10061 Institute of Molecular Cancer Research ,570 Life sciences ,biology ,610 Medicine & health ,2701 Medicine (miscellaneous) ,1100 General Agricultural and Biological Sciences - Published
- 2020
47. Cancer-associated mutations in the iron-sulfur domain of FANCJ affect G-quadruplex metabolism
- Author
-
Odermatt, Diana C, Lee, Wei Ting C, Wild, Sebastian, Jozwiakowski, Stanislaw K, Rothenberg, Eli, Gari, Kerstin, and University of Zurich
- Subjects
10061 Institute of Molecular Cancer Research ,Genetics ,570 Life sciences ,biology ,610 Medicine & health ,QH426-470 - Abstract
FANCJ/BRIP1 is an iron-sulfur (FeS) cluster-binding DNA helicase involved in DNA inter-strand cross-link (ICL) repair and G-quadruplex (G4) metabolism. Mutations in FANCJ are associated with Fanconi anemia and an increased risk for developing breast and ovarian cancer. Several cancer-associated mutations are located in the FeS domain of FANCJ, but how they affect FeS cluster binding and/or FANCJ activity has remained mostly unclear. Here we show that the FeS cluster is indispensable for FANCJ's ability to unwind DNA substrates in vitro and to provide cellular resistance to agents that induce ICLs. Moreover, we find that FANCJ requires an intact FeS cluster for its ability to unfold G4 structures on the DNA template in a primer extension assay with the lagging-strand DNA polymerase delta. Surprisingly, however, FANCJ variants that are unable to bind an FeS cluster and to unwind DNA in vitro can partially suppress the formation of replisome-associated G4 structures that we observe in a FANCJ knock-out cell line. This may suggest a partially retained cellular activity of FANCJ variants with alterations in the FeS domain. On the other hand, FANCJ knock-out cells expressing FeS cluster-deficient variants display a similar-enhanced-sensitivity towards pyridostatin (PDS) and CX-5461, two agents that stabilise G4 structures, as FANCJ knock-out cells. Mutations in FANCJ that abolish FeS cluster binding may hence be predictive of an increased cellular sensitivity towards G4-stabilising agents.
- Published
- 2020
48. DAMEfinder: a method to detect differential allele-specific methylation
- Author
-
Stephany Orjuela, Mark D. Robinson, Dania Machlab, Mirco Menigatti, Giancarlo Marra, and University of Zurich
- Subjects
Epigenomics ,Male ,lcsh:QH426-470 ,Bisulfite sequencing ,610 Medicine & health ,Allele-specific methylation (ASM) ,Single-nucleotide polymorphism ,Biology ,X-inactivation ,Genomic Imprinting ,03 medical and health sciences ,0302 clinical medicine ,1311 Genetics ,1312 Molecular Biology ,Genetics ,Humans ,Epigenetics ,Differential methylation ,Molecular Biology ,Alleles ,030304 developmental biology ,0303 health sciences ,DNA methylation ,Dosage compensation ,10061 Institute of Molecular Cancer Research ,Methodology ,Imprinting ,Sequence Analysis, DNA ,respiratory system ,musculoskeletal system ,respiratory tract diseases ,lcsh:Genetics ,CpG site ,570 Life sciences ,biology ,CpG Islands ,Female ,Colorectal Neoplasms ,Genomic imprinting ,Software ,030217 neurology & neurosurgery - Abstract
Background DNA methylation is a highly studied epigenetic signature that is associated with regulation of gene expression, whereby genes with high levels of promoter methylation are generally repressed. Genomic imprinting occurs when one of the parental alleles is methylated, i.e., when there is inherited allele-specific methylation (ASM). A special case of imprinting occurs during X chromosome inactivation in females, where one of the two X chromosomes is silenced, to achieve dosage compensation between the sexes. Another more widespread form of ASM is sequence dependent (SD-ASM), where ASM is linked to a nearby heterozygous single nucleotide polymorphism (SNP). Results We developed a method to screen for genomic regions that exhibit loss or gain of ASM in samples from two conditions (treatments, diseases, etc.). The method relies on the availability of bisulfite sequencing data from multiple samples of the two conditions. We leverage other established computational methods to screen for these regions within a new R package called DAMEfinder. It calculates an ASM score for all CpG sites or pairs in the genome of each sample, and then quantifies the change in ASM between conditions. It then clusters nearby CpG sites with consistent change into regions. In the absence of SNP information, our method relies only on reads to quantify ASM. This novel ASM score compares favorably to current methods that also screen for ASM. Not only does it easily discern between imprinted and non-imprinted regions, but also females from males based on X chromosome inactivation. We also applied DAMEfinder to a colorectal cancer dataset and observed that colorectal cancer subtypes are distinguishable according to their ASM signature. We also re-discover known cases of loss of imprinting. Conclusion We have designed DAMEfinder to detect regions of differential ASM (DAMEs), which is a more refined definition of differential methylation, and can therefore help in breaking down the complexity of DNA methylation and its influence in development and disease.
- Published
- 2020
- Full Text
- View/download PDF
49. A stapled peptide mimetic of the CtIP tetramerization motif interferes with double-strand break repair and replication fork protection
- Author
-
Christina Zurfluh, Christine von Aesch, Dawid Zyla, Alessandro A. Sartori, R. Scott Williams, Oliver J Wilkinson, Sara Przetocka, Mark S. Dillingham, Jessica L. Wojtaszek, Anika Kuster, Nour L. Mozaffari, University of Zurich, and Sartori, Alessandro A
- Subjects
endocrine system diseases ,DNA damage ,DNA repair ,610 Medicine & health ,Peptide ,Replication fork protection ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,skin and connective tissue diseases ,Research Articles ,030304 developmental biology ,Cancer ,chemistry.chemical_classification ,1000 Multidisciplinary ,0303 health sciences ,Multidisciplinary ,Chemistry ,10061 Institute of Molecular Cancer Research ,SciAdv r-articles ,Cell Biology ,Double Strand Break Repair ,3. Good health ,Chromatin ,Cell biology ,030220 oncology & carcinogenesis ,Cancer cell ,570 Life sciences ,biology ,DNA ,Research Article - Abstract
A stapled peptide targets CtIP tetramers to inhibit DNA repair and exert synthetic lethality in BRCA1-mutant cancer cells., Cancer cells display high levels of DNA damage and replication stress, vulnerabilities that could be exploited by drugs targeting DNA repair proteins. Human CtIP promotes homology-mediated repair of DNA double-strand breaks (DSBs) and protects stalled replication forks from nucleolytic degradation, thus representing an attractive candidate for targeted cancer therapy. Here, we establish a peptide mimetic of the CtIP tetramerization motif that inhibits CtIP activity. The hydrocarbon-stapled peptide encompassing amino acid residues 18 to 28 of CtIP (SP18–28) stably binds to CtIP tetramers in vitro and facilitates their aggregation into higher-order structures. Efficient intracellular uptake of SP18–28 abrogates CtIP localization to damaged chromatin, impairs DSB repair, and triggers extensive fork degradation. Moreover, prolonged SP18–28 treatment causes hypersensitivity to DNA-damaging agents and selectively reduces the viability of BRCA1-mutated cancer cell lines. Together, our data provide a basis for the future development of CtIP-targeting compounds with the potential to treat patients with cancer.
- Published
- 2020
- Full Text
- View/download PDF
50. SMAD1 promoter hypermethylation and lack of SMAD1 expression in Hodgkin lymphoma: a potential target for hypomethylating drug therapy
- Author
-
Visar Vela, Anna Stelling-Germani, Alexandar Tzankov, Claudia Döring, Cheuk Ting Wu, Anne Müller, Sebastian Newrzela, Magdalena M. Gerlach, Sylvia Hartmann, and University of Zurich
- Subjects
business.industry ,10061 Institute of Molecular Cancer Research ,610 Medicine & health ,Hematology ,DNA Methylation ,Hodgkin Disease ,Smad1 Protein ,Pharmacotherapy ,Text mining ,Promoter hypermethylation ,Cancer research ,Humans ,Medicine ,Hodgkin lymphoma ,570 Life sciences ,biology ,Promoter Regions, Genetic ,Letters to the Editor ,business - Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.