1
|
Kaur P, Kumar M, Kaur S, Kumar A, Kaur S. In Vitro Modulation of Genotoxicity and Oxidative Stress by Polyphenol-Rich Fraction of Chinese Ladder Brake (Pteris vittata L.). Appl Biochem Biotechnol 2024; 196:774-789. [PMID: 37195566 DOI: 10.1007/s12010-023-04561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 05/18/2023]
Abstract
Pteris vittata L. is a terrestrial genus growing in moist, shady forests and on hillsides. The plant has considerable ethnomedicinal importance. Investigations have been carried out on chemical profiling and antioxidant compounds from some genera of pteridophytes but studies on the biological properties of P. vittata are lacking. Therefore, the present study investigates antioxidant, antigenotoxic, and antiproliferative potential of the aqueous fraction of P. vittata (PWE). A battery of assays were carried out to assess the antioxidant potential of the PWE. SOS chromotest and DNA nicking assay were used to evaluate the antigenotoxicity of the fraction. The cytotoxic effect of PWE was analyzed using MTT and Neutral Single Cell Gel Electrophoresis comet assay. EC50 of 90.188 µg/ml, 80.13 µg/ml, 142.836 µg/ml, and 12.274 µg/ml was obtained in DPPH, superoxide anion scavenging, reducing power and lipid peroxidation assays, respectively. PWE was potent in inhibiting Fenton's reagent-induced nicking of pBR322 plasmid. The fraction significantly inhibited hydrogen peroxide (H2O2) and 4-nitroquinoline-N-oxide (4NQO) induced mutagenicity and a reduction in induction factor was found with increased PWE concentration. GI50 of 147.16 µg/ml was obtained in MTT assay in human MCF-7 breast cancer cell line. PWE induced apoptosis as confirmed from confocal microscopy studies. The protective effects can be attributed to the presence of the phytochemicals in PWE. These results will be helpful in the development of functional food characteristics, as well as unravel the benefits of pteridophytes as promoters of health.
Collapse
Affiliation(s)
- Paramjeet Kaur
- Genetic Toxicology Laboratory, Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | | | - Sandeep Kaur
- Genetic Toxicology Laboratory, Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Ajay Kumar
- Genetic Toxicology Laboratory, Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
- University Centre for Research & Development (UCRD), Biotechnology Engineering & Food Technology, Chandigarh University, Mohali, India
| | - Satwinderjeet Kaur
- Genetic Toxicology Laboratory, Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
2
|
Rubio-Contreras D, Gómez-Herreros F. TDP1 suppresses chromosomal translocations and cell death induced by abortive TOP1 activity during gene transcription. Nat Commun 2023; 14:6940. [PMID: 37945566 PMCID: PMC10636166 DOI: 10.1038/s41467-023-42622-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
DNA topoisomerase I (TOP1) removes torsional stress by transiently cutting one DNA strand. Such cuts are rejoined by TOP1 but can occasionally become abortive generating permanent protein-linked single strand breaks (SSBs). The repair of these breaks is initiated by tyrosyl-DNA phosphodiesterase 1 (TDP1), a conserved enzyme that unlinks the TOP1 peptide from the DNA break. Additionally, some of these SSBs can result in double strand breaks (DSBs) either during replication or by a poorly understood transcription-associated process. In this study, we identify these DSBs as a source of genome rearrangements, which are suppressed by TDP1. Intriguingly, we also provide a mechanistic explanation for the formation of chromosomal translocations unveiling an error-prone pathway that relies on the MRN complex and canonical non-homologous end-joining. Collectively, these data highlight the threat posed by TOP1-induced DSBs during transcription and demonstrate the importance of TDP1-dependent end-joining in protecting both gene transcription and genome stability.
Collapse
Affiliation(s)
- Diana Rubio-Contreras
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Departamento de Genética, Universidad de Sevilla, 41012, Seville, Spain
| | - Fernando Gómez-Herreros
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.
- Departamento de Genética, Universidad de Sevilla, 41012, Seville, Spain.
| |
Collapse
|
3
|
Frittoli E, Palamidessi A, Iannelli F, Zanardi F, Villa S, Barzaghi L, Abdo H, Cancila V, Beznoussenko GV, Della Chiara G, Pagani M, Malinverno C, Bhattacharya D, Pisati F, Yu W, Galimberti V, Bonizzi G, Martini E, Mironov AA, Gioia U, Ascione F, Li Q, Havas K, Magni S, Lavagnino Z, Pennacchio FA, Maiuri P, Caponi S, Mattarelli M, Martino S, d'Adda di Fagagna F, Rossi C, Lucioni M, Tancredi R, Pedrazzoli P, Vecchione A, Petrini C, Ferrari F, Lanzuolo C, Bertalot G, Nader G, Foiani M, Piel M, Cerbino R, Giavazzi F, Tripodo C, Scita G. Tissue fluidification promotes a cGAS-STING cytosolic DNA response in invasive breast cancer. NATURE MATERIALS 2023; 22:644-655. [PMID: 36581770 PMCID: PMC10156599 DOI: 10.1038/s41563-022-01431-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/02/2022] [Indexed: 05/05/2023]
Abstract
The process in which locally confined epithelial malignancies progressively evolve into invasive cancers is often promoted by unjamming, a phase transition from a solid-like to a liquid-like state, which occurs in various tissues. Whether this tissue-level mechanical transition impacts phenotypes during carcinoma progression remains unclear. Here we report that the large fluctuations in cell density that accompany unjamming result in repeated mechanical deformations of cells and nuclei. This triggers a cellular mechano-protective mechanism involving an increase in nuclear size and rigidity, heterochromatin redistribution and remodelling of the perinuclear actin architecture into actin rings. The chronic strains and stresses associated with unjamming together with the reduction of Lamin B1 levels eventually result in DNA damage and nuclear envelope ruptures, with the release of cytosolic DNA that activates a cGAS-STING (cyclic GMP-AMP synthase-signalling adaptor stimulator of interferon genes)-dependent cytosolic DNA response gene program. This mechanically driven transcriptional rewiring ultimately alters the cell state, with the emergence of malignant traits, including epithelial-to-mesenchymal plasticity phenotypes and chemoresistance in invasive breast carcinoma.
Collapse
Affiliation(s)
| | | | - Fabio Iannelli
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Stefano Villa
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
- Max Plank Institute for Dynamics and Self-Organization, Göttingen, Germany
| | | | - Hind Abdo
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Valeria Cancila
- Department of Health Sciences, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | | | | | - Massimiliano Pagani
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
| | | | | | - Federica Pisati
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Weimiao Yu
- Institute of Molecular and Cell Biology, A*STAR, Singapore, & Bioinformatics Institute, A*STAR, Singapore, Singapore
| | | | | | | | | | - Ubaldo Gioia
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Flora Ascione
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Qingsen Li
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Kristina Havas
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Serena Magni
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Zeno Lavagnino
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Paolo Maiuri
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Silvia Caponi
- Istituto Officina dei Materiali, National Research Council (IOM-CNR), Unit of Perugia, c/o Department of Physics and Geology, University of Perugia, Perugia, Italy
| | | | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, Perugia, Italy
| | - Fabrizio d'Adda di Fagagna
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Institute of Molecular Genetics, National Research Council, Pavia, Italy
| | - Chiara Rossi
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Marco Lucioni
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Richard Tancredi
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- S.C. Oncologia Medica, ASST Melegnano e della Martesana, Ospedale Uboldo, Cernusco sul Naviglio, Milan, Italy
| | - Paolo Pedrazzoli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Roma, La Sapienza, Rome, Italy
| | | | - Francesco Ferrari
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Institute of Molecular Genetics, National Research Council, Pavia, Italy
| | - Chiara Lanzuolo
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
- National Institute of Molecular Genetics Romeo and Enrica Invernizzi, INGM, Milan, Italy
| | - Giovanni Bertalot
- Department of Pathology, S. Chiara Hospital, Azienda Provinciale per i Servizi Sanitari, Trento, Italy
- CISMed University of Trento, University of Trento, Trento, Italy
| | - Guilherme Nader
- Institut Curie and Institut Pierre Gilles de Gennes, PSL Research University, CNRS, UMR-144, Paris, France
- Cell Pathology Children's Hospital of Philadelphia, Research Institute Department of Pathology and Laboratory Medicine University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marco Foiani
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, PSL Research University, CNRS, UMR-144, Paris, France
| | - Roberto Cerbino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
- Faculty of Physics, University of Vienna, Vienna, Austria
| | - Fabio Giavazzi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy.
| | - Claudio Tripodo
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy.
- Department of Health Sciences, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy.
| | - Giorgio Scita
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy.
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
4
|
Bonassi S, Ceppi M, Møller P, Azqueta A, Milić M, Neri M, Brunborg G, Godschalk R, Koppen G, Langie SAS, Teixeira JP, Bruzzone M, Da Silva J, Benedetti D, Cavallo D, Ursini CL, Giovannelli L, Moretti S, Riso P, Del Bo' C, Russo P, Dobrzyńska M, Goroshinskaya IA, Surikova EI, Staruchova M, Barančokova M, Volkovova K, Kažimirova A, Smolkova B, Laffon B, Valdiglesias V, Pastor S, Marcos R, Hernández A, Gajski G, Spremo-Potparević B, Živković L, Boutet-Robinet E, Perdry H, Lebailly P, Perez CL, Basaran N, Nemeth Z, Safar A, Dusinska M, Collins A. DNA damage in circulating leukocytes measured with the comet assay may predict the risk of death. Sci Rep 2021; 11:16793. [PMID: 34408182 PMCID: PMC8373872 DOI: 10.1038/s41598-021-95976-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/02/2021] [Indexed: 01/02/2023] Open
Abstract
The comet assay or single cell gel electrophoresis, is the most common method used to measure strand breaks and a variety of other DNA lesions in human populations. To estimate the risk of overall mortality, mortality by cause, and cancer incidence associated to DNA damage, a cohort of 2,403 healthy individuals (25,978 person-years) screened in 16 laboratories using the comet assay between 1996 and 2016 was followed-up. Kaplan–Meier analysis indicated a worse overall survival in the medium and high tertile of DNA damage (p < 0.001). The effect of DNA damage on survival was modelled according to Cox proportional hazard regression model. The adjusted hazard ratio (HR) was 1.42 (1.06–1.90) for overall mortality, and 1.94 (1.04–3.59) for diseases of the circulatory system in subjects with the highest tertile of DNA damage. The findings of this study provide epidemiological evidence encouraging the implementation of the comet assay in preventive strategies for non-communicable diseases.
Collapse
Affiliation(s)
- Stefano Bonassi
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Rome, Italy. .,Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166, Rome, Italy.
| | - Marcello Ceppi
- Clinical Epidemiology Unit, San Martino Policlinic Hospital, Genoa, Italy
| | - Peter Møller
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Oster Farimagsgade 5A, 1014, Copenhagen, Denmark
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, University of Navarra, C/Irunlarrea 1, 31008, Pamplona, Spain.,C/Irunlarrea 3, IdiSNA, Navarra Institute for Health Research, 31008, Pamplona, Spain
| | - Mirta Milić
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia
| | - Monica Neri
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Rome, Italy
| | - Gunnar Brunborg
- Department of Environmental Health, Section of Molecular Toxicology, Norwegian Institute of Public Health (NIPH), Lovisenberggt 6, 0456, Oslo, Norway
| | - Roger Godschalk
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, University of Maastricht, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - Gudrun Koppen
- Flemish Institute of Technological Research, Environmental Risk and Health Unit VITO - BIOMo, Mol, Belgium
| | - Sabine A S Langie
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, University of Maastricht, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - João Paulo Teixeira
- Environmental Health Department, National Institute of Health, Rua Alexandre Herculano, 321, 4000-055, Porto, Portugal.,Environmental Health Department, Instituto Nacional de Saúde Doutor Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055, Porto, Portugal.,EPIUnit - Instituto de Saúde Pública, Universidade Do Porto, Rua das Taipas, no 135, 4050-600, Porto, Portugal
| | - Marco Bruzzone
- Clinical Epidemiology Unit, San Martino Policlinic Hospital, Genoa, Italy
| | - Juliana Da Silva
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), and La Salle University (UNILASALLE), Canoas, RS, Brazil
| | - Danieli Benedetti
- Laboratory of Genetic Toxicology, Lutheran University of Brazil (ULBRA), and La Salle University (UNILASALLE), Canoas, RS, Brazil
| | - Delia Cavallo
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene (DiMEILA), Italian Workers' Compensation Authority (INAIL), Via Fontana Candida 1, 00078, Monte Porzio Catone (Rome), Italy
| | - Cinzia Lucia Ursini
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene (DiMEILA), Italian Workers' Compensation Authority (INAIL), Via Fontana Candida 1, 00078, Monte Porzio Catone (Rome), Italy
| | - Lisa Giovannelli
- Department NEUROFARBA, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Silvia Moretti
- Department of Health Sciences, Division of Dermatology, University of Florence, Palagi Hospital, Viale Michelangelo 41, Florence, Italy
| | - Patrizia Riso
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133, Milan, Italy
| | - Cristian Del Bo'
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133, Milan, Italy
| | - Patrizia Russo
- Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Rome, Italy.,Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166, Rome, Italy
| | - Malgorzata Dobrzyńska
- Department of Radiation Hygiene and Radiobiology, National Institute of Public Health NIH - National Research Institute, 24 Chocimska Street, 00-791, Warsaw, Poland
| | - Irina A Goroshinskaya
- Laboratory for the Study of the Pathogenesis of Malignant Tumors, National Medical Research Center for Oncology, 14 line 63, 344037, Rostov-on-Don, Russia
| | - Ekaterina I Surikova
- Laboratory for the Study of the Pathogenesis of Malignant Tumors, National Medical Research Center for Oncology, 14 line 63, 344037, Rostov-on-Don, Russia
| | - Marta Staruchova
- Institute of Biology, Medical Faculty, Slovak Medical University, Limbova 12, 83303, Bratislava, Slovakia
| | - Magdalena Barančokova
- Institute of Biology, Medical Faculty, Slovak Medical University, Limbova 12, 83303, Bratislava, Slovakia
| | - Katarina Volkovova
- Institute of Biology, Medical Faculty, Slovak Medical University, Limbova 12, 83303, Bratislava, Slovakia
| | - Alena Kažimirova
- Institute of Biology, Medical Faculty, Slovak Medical University, Limbova 12, 83303, Bratislava, Slovakia
| | - Bozena Smolkova
- Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, Slovakia
| | - Blanca Laffon
- Grupo DICOMOSA, Centro de Investigaciones Científicas Avanzadas (CICA), Departamento de Psicología, Facultad de Ciencias de La Educación, Universidade da Coruña, Campus Elviña s/n, 15071, A Coruña, Spain.,Instituto de Investigación Biomédica de A Coruña (INIBIC), AE CICA-INIBIC, Oza, 15071, A Coruña, Spain
| | - Vanessa Valdiglesias
- Instituto de Investigación Biomédica de A Coruña (INIBIC), AE CICA-INIBIC, Oza, 15071, A Coruña, Spain.,Grupo DICOMOSA, Centro de Investigaciones Científicas Avanzadas (CICA), Departamento de Biología, Facultad de Ciencias, Universidade da Coruña, Campus A Zapateira s/n, 15071, A Coruña, Spain
| | - Susana Pastor
- Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès (Barcelona), Spain
| | - Ricard Marcos
- Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès (Barcelona), Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, 28029, Madrid, Spain
| | - Alba Hernández
- Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès (Barcelona), Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, 28029, Madrid, Spain
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia
| | - Biljana Spremo-Potparević
- Center of Biological Research, Faculty of Pharmacy, University of Belgrade, VojvodeStepe 450, Belgrade, Serbia
| | - Lada Živković
- Center of Biological Research, Faculty of Pharmacy, University of Belgrade, VojvodeStepe 450, Belgrade, Serbia
| | - Elisa Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Pierre Lebailly
- ANTICIPE Unit, INSERM & University of Caen-Normandie Centre François Baclesse, Avenue du Général Harris, 14076, Caen Cedex 05, France
| | - Carlos L Perez
- Department of Biochemistry, Instituto de Ciencias Básicas Y Preclínicas "Victoria de Giron", Universidad de Ciencias Médicas de La Habana, 146 St. and 31 Ave, No, 3102, Playa, Habana, Cuba
| | - Nursen Basaran
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Zsuzsanna Nemeth
- Department of Non-Ionizing Radiation, National Public Health Center, Anna Street 5, 1221, Budapest, Hungary
| | - Anna Safar
- Department of Non-Ionizing Radiation, National Public Health Center, Anna Street 5, 1221, Budapest, Hungary
| | | | - Andrew Collins
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, 0372, Oslo, Norway
| | | |
Collapse
|
5
|
Xu R, Yang Y, Zheng X. Unique structural features of the adenylate kinase hCINAP/AK6 and its multifaceted functions in carcinogenesis and tumor progression. FEBS Lett 2021; 595:2071-2084. [PMID: 34245011 DOI: 10.1002/1873-3468.14158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/30/2022]
Abstract
Human coilin-interacting nuclear ATPase protein (hCINAP), also known as adenylate kinase 6 (AK6), is an atypical adenylate kinase with critical roles in many biological processes, including gene transcription, ribosome synthesis, cell metabolism, cell proliferation and apoptosis, DNA damage responses, and genome stability. Furthermore, hCINAP/AK6 dysfunction is associated with cancer and various inflammatory diseases. In this review, we summarize the structural features and biological roles of hCINAP in several important signaling pathways, as well as its connection with tumor onset and progression.
Collapse
Affiliation(s)
- Ruidan Xu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Yongfeng Yang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Xiaofeng Zheng
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
6
|
Qiu TA, Guidolin V, Hoang KNL, Pho T, Carra' A, Villalta PW, He J, Yao X, Hamers RJ, Balbo S, Feng ZV, Haynes CL. Nanoscale battery cathode materials induce DNA damage in bacteria. Chem Sci 2020; 11:11244-11258. [PMID: 34094365 PMCID: PMC8162401 DOI: 10.1039/d0sc02987d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/19/2020] [Indexed: 11/21/2022] Open
Abstract
The increasing use of nanoscale lithium nickel manganese cobalt oxide (Li x Ni y Mn z Co1-y-z O2, NMC) as a cathode material in lithium-ion batteries poses risk to the environment. Learning toxicity mechanisms on molecular levels is critical to promote proactive risk assessment of these complex nanomaterials and inform their sustainable development. We focused on DNA damage as a toxicity mechanism and profiled in depth chemical and biological changes linked to DNA damage in two environmentally relevant bacteria upon nano-NMC exposure. DNA damage occurred in both bacteria, characterized by double-strand breakage and increased levels of many putative chemical modifications on bacterial DNA bases related to direct oxidative stress and lipid peroxidation, measured by cutting-edge DNA adductomic techniques. Chemical probes indicated elevated intracellular reactive oxygen species and transition metal ions, in agreement with DNA adductomics and gene expression analysis. By integrating multi-dimensional datasets from chemical and biological measurements, we present rich mechanistic insights on nano-NMC-induced DNA damage in bacteria, providing targets for biomarkers in the risk assessment of reactive materials that may be extrapolated to other nano-bio interactions.
Collapse
Affiliation(s)
- Tian A Qiu
- Department of Chemistry, University of Minnesota 207 Pleasant St SE Minneapolis MN 55455 USA
| | - Valeria Guidolin
- Masonic Cancer Center, University of Minnesota 2231 6th Street SE Minneapolis MN 55455 USA
| | - Khoi Nguyen L Hoang
- Chemistry Department, Augsburg University 2211 Riverside Ave Minneapolis MN 55454 USA
| | - Thomas Pho
- Chemistry Department, Augsburg University 2211 Riverside Ave Minneapolis MN 55454 USA
| | - Andrea Carra'
- Masonic Cancer Center, University of Minnesota 2231 6th Street SE Minneapolis MN 55455 USA
| | - Peter W Villalta
- Masonic Cancer Center, University of Minnesota 2231 6th Street SE Minneapolis MN 55455 USA
| | - Jiayi He
- Department of Chemistry, University of Minnesota 207 Pleasant St SE Minneapolis MN 55455 USA
| | - Xiaoxiao Yao
- Department of Chemistry, University of Minnesota 207 Pleasant St SE Minneapolis MN 55455 USA
| | - Robert J Hamers
- Department of Chemistry, University of Wisconsin 1101 University Avenue Madison WI 53706 USA
| | - Silvia Balbo
- Masonic Cancer Center, University of Minnesota 2231 6th Street SE Minneapolis MN 55455 USA
| | - Z Vivian Feng
- Chemistry Department, Augsburg University 2211 Riverside Ave Minneapolis MN 55454 USA
| | - Christy L Haynes
- Department of Chemistry, University of Minnesota 207 Pleasant St SE Minneapolis MN 55455 USA
| |
Collapse
|
7
|
Pepoyan AZ, Balayan MH, Malkhasyan L, Manvelyan A, Bezhanyan T, Paronikyan R, Tsaturyan VV, Tatikyan S, Kamiya S, Chikindas ML. Effects of Probiotic Lactobacillus acidophilus Strain INMIA 9602 Er 317/402 and Putative Probiotic Lactobacilli on DNA Damages in the Small Intestine of Wistar Rats In Vivo. Probiotics Antimicrob Proteins 2020; 11:905-909. [PMID: 30515721 DOI: 10.1007/s12602-018-9491-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Double-strand breaks in the DNA of the small intestine in male Wistar rats were studied using a neutral comet assay after 7 days of feeding with a single strain probiotic formulation Narine (Vitamax-E, Armenia), containing Lactobacillus acidophilus strain Er-2317/402 Narine, and putative probiotics L. rhamnosus Vahe and L. delbrueckii IAHAHI. Type 0 (undamaged DNA), type 1 (head diameter 13.18-17.08 μm), and type 2 (14.15-μm head diameter) damaged DNA comets were studied in control and lactobacilli-fed rats using the neutral comet assay. Lactobacilli-fed rats were shown to carry only type 0 (undamaged) DNA.Thus, the effects of probiotic Lactobacillus acidophilus strain INMIA 9602 Er 317/402 and putative probiotic lactobacilli on DNA damage in the small intestine of Wistar rats in vivo was shown, and the neutral comet assay is suggested as a potential tool for the in vivo selection of putative probiotics with DNA-protective activity.
Collapse
Affiliation(s)
- Astghik Z Pepoyan
- Department of Food Safety and Biotechnology, Armenian National Agrarian University, Teryan 74, 0009, Yerevan, Armenia.
| | - Marine H Balayan
- International Association for Human and Animals Health Improvement, Azatutyan 11, 0037, Yerevan, Armenia
| | - Lilit Malkhasyan
- International Association for Human and Animals Health Improvement, Azatutyan 11, 0037, Yerevan, Armenia
| | - Anahit Manvelyan
- Department of Food Safety and Biotechnology, Armenian National Agrarian University, Teryan 74, 0009, Yerevan, Armenia
| | - Tatevik Bezhanyan
- International Association for Human and Animals Health Improvement, Azatutyan 11, 0037, Yerevan, Armenia
| | - Ruzanna Paronikyan
- International Association for Human and Animals Health Improvement, Azatutyan 11, 0037, Yerevan, Armenia
| | | | - Stepan Tatikyan
- "CANDLE" Synchrotron Research Institute, 0037, Yerevan, Armenia
| | - Shigeru Kamiya
- Kyorin University School of Medicine, Tokyo, 192-0005, Japan
| | - Michael L Chikindas
- Health Promoting Natural Laboratory, Rutgers State University, New Brunswick, NJ, 08901, USA
| |
Collapse
|
8
|
Clementi E, Inglin L, Beebe E, Gsell C, Garajova Z, Markkanen E. Persistent DNA damage triggers activation of the integrated stress response to promote cell survival under nutrient restriction. BMC Biol 2020; 18:36. [PMID: 32228693 PMCID: PMC7106853 DOI: 10.1186/s12915-020-00771-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Base-excision repair (BER) is a central DNA repair mechanism responsible for the maintenance of genome integrity. Accordingly, BER defects have been implicated in cancer, presumably by precipitating cellular transformation through an increase in the occurrence of mutations. Hence, tight adaptation of BER capacity is essential for DNA stability. However, counterintuitive to this, prolonged exposure of cells to pro-inflammatory molecules or DNA-damaging agents causes a BER deficiency by downregulating the central scaffold protein XRCC1. The rationale for this XRCC1 downregulation in response to persistent DNA damage remains enigmatic. Based on our previous findings that XRCC1 downregulation causes wide-ranging anabolic changes, we hypothesised that BER depletion could enhance cellular survival under stress, such as nutrient restriction. RESULTS Here, we demonstrate that persistent single-strand breaks (SSBs) caused by XRCC1 downregulation trigger the integrated stress response (ISR) to promote cellular survival under nutrient-restricted conditions. ISR activation depends on DNA damage signalling via ATM, which triggers PERK-mediated eIF2α phosphorylation, increasing translation of the stress-response factor ATF4. Furthermore, we demonstrate that SSBs, induced either through depletion of the transcription factor Sp1, responsible for XRCC1 levels, or through prolonged oxidative stress, trigger ISR-mediated cell survival under nutrient restriction as well. Finally, the ISR pathway can also be initiated by persistent DNA double-strand breaks. CONCLUSIONS Our results uncover a previously unappreciated connection between persistent DNA damage, caused by a decrease in BER capacity or direct induction of DNA damage, and the ISR pathway that supports cell survival in response to genotoxic stress with implications for tumour biology and beyond.
Collapse
Affiliation(s)
- Elena Clementi
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Larissa Inglin
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Corina Gsell
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Zuzana Garajova
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland.
| |
Collapse
|
9
|
Matsui A, Kobayashi J, Kanno SI, Hashiguchi K, Miyaji M, Yoshikawa Y, Yasui A, Zhang-Akiyama QM. Oxidation resistance 1 prevents genome instability through maintenance of G2/M arrest in gamma-ray-irradiated cells. JOURNAL OF RADIATION RESEARCH 2020; 61:1-13. [PMID: 31845986 PMCID: PMC6976731 DOI: 10.1093/jrr/rrz080] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 09/18/2019] [Indexed: 06/10/2023]
Abstract
Human oxidation resistance 1 (OXR1) was identified as a protein that decreases genomic mutations in Escherichia coli caused by oxidative DNA damage. However, the mechanism by which OXR1 defends against genome instability has not been elucidated. To clarify how OXR1 maintains genome stability, the effects of OXR1-depletion on genome stability were investigated in OXR1-depleted HeLa cells using gamma-rays (γ-rays). The OXR1-depleted cells had higher levels of superoxide and micronucleus (MN) formation than control cells after irradiation. OXR1-overexpression alleviated the increases in reactive oxygen species (ROS) level and MN formation after irradiation. The increased MN formation in irradiated OXR1-depleted cells was partially attenuated by the ROS inhibitor N-acetyl-L-cysteine, suggesting that OXR1-depeletion increases ROS-dependent genome instability. We also found that OXR1-depletion shortened the duration of γ-ray-induced G2/M arrest. In the presence of the cell cycle checkpoint inhibitor caffeine, the level of MN formed after irradiation was similar between control and OXR1-depleted cells, demonstrating that OXR1-depletion accelerates MN formation through abrogation of G2/M arrest. In OXR1-depleted cells, the level of cyclin D1 protein expression was increased. Here we report that OXR1 prevents genome instability by cell cycle regulation as well as oxidative stress defense.
Collapse
Affiliation(s)
- Ako Matsui
- Laboratory of Stress Response Biology, Department of Zoology, Division of Biological Sciences, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Junya Kobayashi
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shin-ichiro Kanno
- Division of Dynamic Proteome in Cancer and Aging, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryocho, Aobaku, Sendai 980-8575, Japan
| | - Kazunari Hashiguchi
- Laboratory of Stress Response Biology, Department of Zoology, Division of Biological Sciences, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
- Department of Biochemistry, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Masahiro Miyaji
- Laboratory of Stress Response Biology, Department of Zoology, Division of Biological Sciences, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yukihiro Yoshikawa
- Laboratory of Stress Response Biology, Department of Zoology, Division of Biological Sciences, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Akira Yasui
- Division of Dynamic Proteome in Cancer and Aging, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryocho, Aobaku, Sendai 980-8575, Japan
| | - Qiu-Mei Zhang-Akiyama
- Laboratory of Stress Response Biology, Department of Zoology, Division of Biological Sciences, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
10
|
Codrich M, Comelli M, Malfatti MC, Mio C, Ayyildiz D, Zhang C, Kelley MR, Terrosu G, Pucillo CEM, Tell G. Inhibition of APE1-endonuclease activity affects cell metabolism in colon cancer cells via a p53-dependent pathway. DNA Repair (Amst) 2019; 82:102675. [PMID: 31450087 PMCID: PMC7092503 DOI: 10.1016/j.dnarep.2019.102675] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
The pathogenesis of colorectal cancer (CRC) involves different mechanisms, such as genomic and microsatellite instabilities. Recently, a contribution of the base excision repair (BER) pathway in CRC pathology has been emerged. In this context, the involvement of APE1 in the BER pathway and in the transcriptional regulation of genes implicated in tumor progression strongly correlates with chemoresistance in CRC and in more aggressive cancers. In addition, the APE1 interactome is emerging as an important player in tumor progression, as demonstrated by its interaction with Nucleophosmin (NPM1). For these reasons, APE1 is becoming a promising target in cancer therapy and a powerful prognostic and predictive factor in several cancer types. Thus, specific APE1 inhibitors have been developed targeting: i) the endonuclease activity; ii) the redox function and iii) the APE1-NPM1 interaction. Furthermore, mutated p53 is a common feature of advanced CRC. The relationship between APE1 inhibition and p53 is still completely unknown. Here, we demonstrated that the inhibition of the endonuclease activity of APE1 triggers p53-mediated effects on cell metabolism in HCT-116 colon cancer cell line. In particular, the inhibition of the endonuclease activity, but not of the redox function or of the interaction with NPM1, promotes p53 activation in parallel to sensitization of p53-expressing HCT-116 cell line to genotoxic treatment. Moreover, the endonuclease inhibitor affects mitochondrial activity in a p53-dependent manner. Finally, we demonstrated that 3D organoids derived from CRC patients are susceptible to APE1-endonuclease inhibition in a p53-status correlated manner, recapitulating data obtained with HCT-116 isogenic cell lines. These findings suggest the importance of further studies aimed at testing the possibility to target the endonuclease activity of APE1 in CRC.
Collapse
Affiliation(s)
- Marta Codrich
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Marina Comelli
- Laboratory of Bioenergetics, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Matilde Clarissa Malfatti
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Catia Mio
- Institute of Medical Genetics, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Dilara Ayyildiz
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Mark R Kelley
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and Pharmacology & Toxicology, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Giovanni Terrosu
- General Surgery and Transplantation Unit, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Carlo E M Pucillo
- Laboratory of Immunology, Department of Medicine, University of Udine, Udine, 33100, Italy
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, Udine, 33100, Italy.
| |
Collapse
|
11
|
McKay MJ, Craig J, Kalitsis P, Kozlov S, Verschoor S, Chen P, Lobachevsky P, Vasireddy R, Yan Y, Ryan J, McGillivray G, Savarirayan R, Lavin MF, Ramsay RG, Xu H. A Roberts Syndrome Individual With Differential Genotoxin Sensitivity and a DNA Damage Response Defect. Int J Radiat Oncol Biol Phys 2019; 103:1194-1202. [PMID: 30508616 DOI: 10.1016/j.ijrobp.2018.11.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/14/2018] [Accepted: 11/23/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE Roberts syndrome (RBS) is a rare, recessively transmitted developmental disorder characterized by growth retardation, craniofacial abnormalities, and truncation of limbs. All affected individuals to date have mutations in the ESCO2 (establishment of cohesion 2) gene, a key regulator of the cohesin complex, which is involved in sister chromatid cohesion and DNA double-strand break (DSB) repair. Here we characterize DNA damage responses (DDRs) for the first time in an RBS-affected family. METHODS AND MATERIALS Lymphoblastoid cell lines were established from an RBS family, including the proband and parents carrying ESCO2 mutations. Various DDR assays were performed on these cells, including cell survival, chromosome break, and apoptosis assays; checkpoint activation indicators; and measures of DNA breakage and repair. RESULTS Cells derived from the RBS-affected individual showed sensitivity to ionizing radiation (IR) and mitomycin C-induced DNA damage. In this ESCO2 compound heterozygote, other DDRs were also defective, including enhanced IR-induced clastogenicity and apoptosis; increased DNA DSB induction; and a reduced capacity for repairing IR-induced DNA DSBs, as measured by γ-H2AX foci and the comet assay. CONCLUSIONS In addition to its developmental features, RBS can be, like ataxia telangiectasia, considered a DDR-defective syndrome, which contributes to its cellular, molecular, and clinical phenotype.
Collapse
Affiliation(s)
- Michael J McKay
- Olivia Newton-John Cancer Research Institute and Austin Health, Heidelberg, Victoria, Australia; Latrobe University, Bundoora, Victoria, Australia
| | - Jeffery Craig
- School of Medicine, Deakin University, Geelong Waurn Campus, Geelong, Victoria, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Paul Kalitsis
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Sergei Kozlov
- University of Queensland Centre for Clinical Research, Royal Brisbane & Women's Hospital Campus, Herston, Queensland, Australia
| | - Sandra Verschoor
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Phillip Chen
- University of Queensland Centre for Clinical Research, Royal Brisbane & Women's Hospital Campus, Herston, Queensland, Australia
| | - Pavel Lobachevsky
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Raja Vasireddy
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Yuqian Yan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jacinta Ryan
- School of Medicine, Flinders University, Adelaide, South Australia, Australia; Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - George McGillivray
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Ravi Savarirayan
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Martin F Lavin
- University of Queensland Centre for Clinical Research, Royal Brisbane & Women's Hospital Campus, Herston, Queensland, Australia
| | - Robert G Ramsay
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Huiling Xu
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Clinical Pathology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia; College of Life Sciences, Shanxi Normal University, Linfen, Shanxi, China.
| |
Collapse
|
12
|
Kaur V, Kumar M, Kumar A, Kaur S. Butea monosperma (Lam.) Taub. Bark fractions protect against free radicals and induce apoptosis in MCF-7 breast cancer cells via cell-cycle arrest and ROS-mediated pathway. Drug Chem Toxicol 2018; 43:398-408. [DOI: 10.1080/01480545.2018.1497051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Varinder Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, India
| | - Manish Kumar
- Department of Biology, S.D. College, Barnala, India
| | - Ajay Kumar
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, India
| | - Satwinderjeet Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
13
|
Jafari N, Giehr P, Hesaraki M, Baas R, de Graaf P, Timmers HTM, Walter J, Baharvand H, Totonchi M. Genomic integrity of ground-state pluripotency. J Cell Biochem 2018; 119:9781-9789. [PMID: 30171711 DOI: 10.1002/jcb.27296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/25/2018] [Indexed: 11/06/2022]
Abstract
Pluripotent cells appear to be in a transient state during early development. These cells have the capability to transition into embryonic stem cells (ESCs). It has been reported that mouse pluripotent cells cultivated in chemically defined media sustain the ground state of pluripotency. Because the epigenetic pattern of pluripotent cells reflects their environment, culture under different conditions causes epigenetic changes, which could lead to genomic instability. This study focused on the DNA methylation pattern of repetitive elements (REs) and their activation levels under two ground-state conditions and assessed the genomic integrity of ESCs. We measured the methylation and expression level of REs in different media. The results indicated that although the ground-state conditions show higher REs activity, they did not lead to DNA damage; therefore, the level of genomic instability is lower under the ground-state compared with the conventional condition. Our results indicated that when choosing an optimum condition, different features of the condition must be considered to have epigenetically and genomically stable stem cells.
Collapse
Affiliation(s)
- Narges Jafari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Pascal Giehr
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Roy Baas
- Regenerative Medicine Center and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Petra de Graaf
- Regenerative Medicine Center and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H T Marc Timmers
- Regenerative Medicine Center and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Urology, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), University Medical Center Freiburg, Freiburg, Germany
| | - Jörn Walter
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
14
|
Vasconcelos ZS, Ralph ACL, Calcagno DQ, dos Santos Barbosa G, do Nascimento Pedrosa T, Antony LP, de Arruda Cardoso Smith M, de Lucas Chazin E, Vasconcelos TRA, Montenegro RC, de Vasconcellos MC. Anticancer potential of benzothiazolic derivative (E)-2-((2-(benzo[d]thiazol-2-yl)hydrazono)methyl)-4-nitrophenol against melanoma cells. Toxicol In Vitro 2018; 50:225-235. [DOI: 10.1016/j.tiv.2018.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 02/22/2018] [Accepted: 03/01/2018] [Indexed: 10/17/2022]
|
15
|
Intrinsic toxicity of stable nanosized titanium dioxide using polyacrylate in human keratinocytes. Mol Cell Toxicol 2018. [DOI: 10.1007/s13273-018-0030-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
16
|
Damiani RM, Moura DJ, Viau CM, Brito V, Morás AM, Henriques JAP, Saffi J. Influence of PARP-1 inhibition in the cardiotoxicity of the topoisomerase 2 inhibitors doxorubicin and mitoxantrone. Toxicol In Vitro 2018; 52:203-213. [PMID: 29913208 DOI: 10.1016/j.tiv.2018.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/17/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) and Mitoxantrone (MTX) are very effective drugs for a range of tumors despite being highly cardiotoxic. DNA topoisomerase 2 beta (Top2ß) was revealed as key mediator of DOX-induced cardiotoxicity, although ROS generation is also an important mechanism. Oxidative stress is also an important issue in MTX-induced cardiotoxicity that is manifested by mitochondrial dysfunction. Studies have demonstrated the relationship between PARP-1 overactivation and cell viability in DOX-treated cardiomyocytes. In reference of MTX, data regarding PARP-1 overactivation as the mechanism responsible for cardiotoxicity is difficult to find. The aim of this study was to evaluate the influence of PARP-1 inhibitor DPQ on DOX- and MTX-mediated cardiotoxicity. Cells were exposed for 24 h to DOX or MTX in the presence or absence of DPQ. Viability, apoptosis, and genotoxicity assays were carried out. Immunofluorescence of phosphorylated histone H2AX was analyzed in H9c2 cells and cardiomyocytes from neonatal rats. Results demonstrated that DPQ co-treatment increases DOX-induced apoptosis in H9c2 cells. DPQ also prevents DOX and MTX-ROS generation in part by increasing SOD and CAT activities. Furthermore, DPQ co-treatment increased the generation of DNA strand breaks by DOX and MTX whilst also inducing phosphorylation of H2AX, MRE11, and ATM in H9c2 cells. Our results demonstrated that as well as increasing DNA damage and inducing apoptotic cell death, DPQ enhances DOX- and MTX-mediated cytotoxicity in H9c2.
Collapse
Affiliation(s)
- Roberto Marques Damiani
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil; Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil; Centro Universitário Ritter dos Reis (UniRitter), Orfanotrófio st, 555, Porto Alegre, RS, Brazil.
| | - Dinara Jaqueline Moura
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - Cassiana Macagnan Viau
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - Verônica Brito
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - Ana Moira Morás
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil
| | - João Antonio Pêgas Henriques
- Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil; Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil
| | - Jenifer Saffi
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite st., 245, Porto Alegre, RS, Brazil; Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves av., 9500, Porto Alegre, RS, Brazil
| |
Collapse
|
17
|
Mórocz M, Zsigmond E, Tóth R, Enyedi MZ, Pintér L, Haracska L. DNA-dependent protease activity of human Spartan facilitates replication of DNA-protein crosslink-containing DNA. Nucleic Acids Res 2017; 45:3172-3188. [PMID: 28053116 PMCID: PMC5389635 DOI: 10.1093/nar/gkw1315] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/22/2016] [Indexed: 01/24/2023] Open
Abstract
Mutations in SPARTAN are associated with early onset hepatocellular carcinoma and progeroid features. A regulatory function of Spartan has been implicated in DNA damage tolerance pathways such as translesion synthesis, but the exact function of the protein remained unclear. Here, we reveal the role of human Spartan in facilitating replication of DNA–protein crosslink-containing DNA. We found that purified Spartan has a DNA-dependent protease activity degrading certain proteins bound to DNA. In concert, Spartan is required for direct DPC removal in vivo; we also show that the protease Spartan facilitates repair of formaldehyde-induced DNA–protein crosslinks in later phases of replication using the bromodeoxyuridin (BrdU) comet assay. Moreover, DNA fibre assay indicates that formaldehyde-induced replication stress dramatically decreases the speed of replication fork movement in Spartan-deficient cells, which accumulate in the G2/M cell cycle phase. Finally, epistasis analysis mapped these Spartan functions to the RAD6-RAD18 DNA damage tolerance pathway. Our results reveal that Spartan facilitates replication of DNA–protein crosslink-containing DNA enzymatically, as a protease, which may explain its role in preventing carcinogenesis and aging.
Collapse
Affiliation(s)
- Mónika Mórocz
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Eszter Zsigmond
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Róbert Tóth
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Márton Zs Enyedi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Lajos Pintér
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| | - Lajos Haracska
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6726, Hungary
| |
Collapse
|
18
|
PLGA-CTAB curcumin nanoparticles: Fabrication, characterization and molecular basis of anticancer activity in triple negative breast cancer cell lines (MDA-MB-231 cells). Biomed Pharmacother 2017; 94:944-954. [PMID: 28810532 DOI: 10.1016/j.biopha.2017.07.151] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/16/2017] [Accepted: 07/30/2017] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancers (TNBC) are aggressive cancers, which do not control by hormonal therapy or therapies that target HER-2 receptors. Curcumin (Cur) has shown cytotoxic effects in multiple cancer cell lines. However, its medical uses remain limited due to low aqueous solubility and poor bioavailability. Therefore, present study was aimed to fabricate the small positive charge curcumin nanoparticles (CN) by nanoprecipitation methods using PLGA and CTAB, and to evaluate its anticancer efficacy and underlying the mechanism in triple negative breast cancer cell lines (MDA-MB-231 cells). In in-vitro drug release assay, Cur was released from CN by flicking diffusion and anomalous transport process. CN showed a higher cellular incorporation than free Cur resulted in higher cytotoxicity. Checking the anticancer activity at the molecular level, Cur has shown to induce the reactive oxygen species production that subsequently causes the DNA damage and resulting in p38-MAPK activation. The p38-MAPK induce the expression of p16/INKK4a, p21/waf1/cip1 and p53 resulting in a reduction in the level of CDK2, CDK4, cyclin D1 and cyclin E and subsequently cell cycle arrest at G1/S and G2/M phase. It also reduces the expression of DNA repair gene, i.e. BRCA1, BRCA2, Rad51, Rad50, Mre11 and NBS1 resulting in apoptosis induction due to persistent DNA damage. This study presents an effective delivery of curcumin in TNBC cancer cells and it could open the new frontiers in clinical cancer chemotherapy.
Collapse
|
19
|
Torres TE, Russo LC, Santos A, Marques GR, Magalhaes YT, Tabassum S, Forti FL. Loss of DUSP3 activity radiosensitizes human tumor cell lines via attenuation of DNA repair pathways. Biochim Biophys Acta Gen Subj 2017; 1861:1879-1894. [PMID: 28389334 DOI: 10.1016/j.bbagen.2017.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/22/2017] [Accepted: 04/02/2017] [Indexed: 12/19/2022]
|
20
|
Kaur V, Kumar M, Kaur P, Kaur S, Kaur S. Inhibitory Activities of Butanol Fraction from Butea monosperma
(Lam
.) Taub
. Bark Against Free Radicals, Genotoxins and Cancer Cells. Chem Biodivers 2017; 14. [DOI: 10.1002/cbdv.201600484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/13/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Varinder Kaur
- Department of Botanical and Environmental Sciences; Guru Nanak Dev University; Amritsar Punjab 143005 India
| | - Manish Kumar
- Department of Botanical and Environmental Sciences; Guru Nanak Dev University; Amritsar Punjab 143005 India
- Akal College of Basic Sciences (Botany); Eternal University; Baru Sahib Sirmour Himachal Pradesh 173101 India
| | - Paramjeet Kaur
- Department of Botanical and Environmental Sciences; Guru Nanak Dev University; Amritsar Punjab 143005 India
| | - Sandeep Kaur
- Department of Botanical and Environmental Sciences; Guru Nanak Dev University; Amritsar Punjab 143005 India
| | - Satwinderjeet Kaur
- Department of Botanical and Environmental Sciences; Guru Nanak Dev University; Amritsar Punjab 143005 India
| |
Collapse
|
21
|
Xiao M, Cai J, Cai L, Jia J, Xie L, Zhu Y, Huang B, Jin D, Wang Z. Let-7e sensitizes epithelial ovarian cancer to cisplatin through repressing DNA double strand break repair. J Ovarian Res 2017; 10:24. [PMID: 28376831 PMCID: PMC5379542 DOI: 10.1186/s13048-017-0321-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/28/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Resistance to platinum-based chemotherapy remains a great challenge for ovarian cancer treatment. The human let-7 family contains 13 members located on nine different chromosomes, and most members have been implicated in the modulation of drug sensitivity in cancers. Our previous study showed that deregulation of let-7e in epithelial ovarian cancer (EOC) promoted the development of resistance to cisplatin. In the present study, we aimed to investigate the underlying mechanism and further evaluate the clinical value of let-7e in predicting chemo-response and prognosis in EOC. RESULTS In situ hybridization assays revealed a significantly decreased expression of let-7e in chemo-resistant EOC tissues compared with chemo-sensitive cases. Transfection with let-7e agomir sensitized EOC cells to cisplatin, down-regulated BRCA1 and Rad51 expression, and repressed the repair of cisplatin-induced DNA double strand break, while let-7e inhibitor exerted the opposite effects. In human EOC tissues, BRCA1 and Rad51 levels were increased in the chemo-resistant group compared with the sensitive group and were negatively correlated with let-7e. Low let-7e and high Rad51 were significantly associated with poor progression-free survival and overall survival and multivariate regression analyses showed that let-7e was an independent predictor for overall survival and chemotherapy response in EOC. Receiver operating characteristic analysis indicated that let-7e level was highly predictive of resistance to platinum-taxane chemotherapy with an area under the curve of 0.826. CONCLUSIONS In EOC, low let-7e leads to activation of BRCA1 and Rad51 expression and subsequent enhancement of DSB repair, which in turn results in cisplatin-resistance. Let-7e is a potential predictor for survival and chemo-response in EOC and re-expression of let-7e might be an effective strategy for overcoming chemo-resistance.
Collapse
Affiliation(s)
- Man Xiao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Liqiong Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jinghui Jia
- Department of Obstetrics and Gynecology, Air Force General Hospital, PLA, Beijing, 100142, China
| | - Lisha Xie
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Ying Zhu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Bangxing Huang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Dongdong Jin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
22
|
SNEV hPrp19/hPso4 Regulates Adipogenesis of Human Adipose Stromal Cells. Stem Cell Reports 2016; 8:21-29. [PMID: 28041875 PMCID: PMC5233435 DOI: 10.1016/j.stemcr.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 11/26/2022] Open
Abstract
Aging is accompanied by loss of subcutaneous adipose tissue. This may be due to reduced differentiation capacity or deficiency in DNA damage repair (DDR) factors. Here we investigated the role of SNEVhPrp19/hPso4, which was implicated in DDR and senescence evasion, in adipogenic differentiation of human adipose stromal cells (hASCs). We showed that SNEV is induced during adipogenesis and localized both in the nucleus and in the cytoplasm. Knockdown of SNEV perturbed adipogenic differentiation and led to accumulation of DNA damage in hASCs upon oxidative stress. In addition, we demonstrated that SNEV is required for fat deposition in Caenorhabditis elegans. Consequently, we tested other DDR factors and found that WRN is also required for adipogenesis in both models. These results demonstrate that SNEV regulates adipogenesis in hASCs and indicate that DDR capacity in general might be a pre-requisite for this process. SNEV is required for adipogenic differentiation of human adipose stromal cells SNEV modulates pro- and anti-adipogenic signaling pathways SNEV regulates DNA repair capacity of human adipose stromal cells SNEV modulates organismal fat deposition in Caenorhabditis elegans
Collapse
|
23
|
Ralph ACL, Calcagno DQ, da Silva Souza LG, de Lemos TLG, Montenegro RC, de Arruda Cardoso Smith M, de Vasconcellos MC. Biflorin induces cytotoxicity by DNA interaction in genetically different human melanoma cell lines. Toxicol In Vitro 2016; 34:237-245. [DOI: 10.1016/j.tiv.2016.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/08/2016] [Accepted: 04/08/2016] [Indexed: 11/24/2022]
|
24
|
Fu HC, Yang YC, Chen YJ, Lin H, Ou YC, Chien CCC, Huang EY, Huang HY, Lan J, Chi HP, Huang KE, Kang HY. Increased expression of SKP2 is an independent predictor of locoregional recurrence in cervical cancer via promoting DNA-damage response after irradiation. Oncotarget 2016; 7:44047-44061. [PMID: 27317767 PMCID: PMC5190078 DOI: 10.18632/oncotarget.10057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/16/2016] [Indexed: 11/25/2022] Open
Abstract
Although radiation therapy was known to be effective to cervical cancer, loco-regional recurrences are frequently found in patients. We aimed to identify a molecular marker predicting the response of cervical cancer to radiotherapy. We included the patients (n = 149) with cervical cancer who had undergone radiotherapy from 2004 to 2006. Tumor samples were collected to examine the association between the expression of S-phase kinase-associated protein 2 (SKP2) and prognosis in cervical cancer. We found higher expression of SKP2 associated with recurrence (HRs: 2.52, p < 0.001), death (HRs: 2.01, p < 0.001) and higher locoregional recurrence rate (HRs: 3.76, p < 0.001). Cervical cancer cell lines with higher expression of SKP2 showed higher colony formation, cell survival rate and fewer DNA damages after irradiation. SKP2-C25, an inhibitor for SKP2 activity, dose-dependently decreased cell viability after irradiation and knockdown of SKP2 impaired DNA-damage response and sensitized the cervical cancer cells to irradiation. Our data showed the SKP2 represents a promising tool to identify patients with cervical cancer who have a higher risk of locoregional recurrence after radiotherapy. Targeting SKP2 may serve as a potential radiosensitizer for developing effective therapeutic strategies against cervical cancer.
Collapse
Affiliation(s)
- Hung-Chun Fu
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chien Yang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yun-Ju Chen
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chan-Chao Chang Chien
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Eng-Yen Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jui Lan
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsi-Ping Chi
- Medical Sciences Division, University of Oxford, Oxford, England, UK
| | - Ko-En Huang
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
25
|
Huang Y, Wang X, Niu X, Wang X, Jiang R, Xu T, Liu Y, Liang L, Ou X, Xing X, Li W, Hu C. EZH2 suppresses the nucleotide excision repair in nasopharyngeal carcinoma by silencing XPA gene. Mol Carcinog 2016; 56:447-463. [DOI: 10.1002/mc.22507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 05/05/2016] [Accepted: 05/31/2016] [Indexed: 12/24/2022]
Affiliation(s)
- Yuxiang Huang
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Xuanyi Wang
- Institute of Traditional Chinese Medicine and Western Medicine; School of Medicine; Yangzhou University; Yangzhou China
| | - Xiaoshuang Niu
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Xiaoshen Wang
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Rui Jiang
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Tingting Xu
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Yong Liu
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Liping Liang
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Xiaomin Ou
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Xing Xing
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Weiwei Li
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Chaosu Hu
- Department of Radiation Oncology; Fudan University Shanghai Cancer Center; Shanghai China
| |
Collapse
|
26
|
Rocca CJ, Soares DG, Bouzid H, Henriques JAP, Larsen AK, Escargueil AE. BRCA2 is needed for both repair and cell cycle arrest in mammalian cells exposed to S23906, an anticancer monofunctional DNA binder. Cell Cycle 2016; 14:2080-90. [PMID: 25945522 DOI: 10.1080/15384101.2015.1042632] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Repair of DNA-targeted anticancer agents is an active area of investigation of both fundamental and clinical interest. However, most studies have focused on a small number of compounds limiting our understanding of both DNA repair and the DNA damage response. S23906 is an acronycine derivative that shows strong activity toward solid tumors in experimental models. S23906 forms bulky monofunctional DNA adducts in the minor groove which leads to destabilization of the double-stranded helix. We now report that S23906 induces formation of DNA double strand breaks that are processed through homologous recombination (HR) but not Non-Homologous End-Joining (NHEJ) repair. Interestingly, S23906 exposure was accompanied by a higher sensitivity of BRCA2-deficient cells compared to other HR deficient cell lines and by an S-phase accumulation in wild-type (wt), but not in BRCA2-deficient cells. Recently, we have shown that S23906-induced S phase arrest was mediated by the checkpoint kinase Chk1. However, its activated phosphorylated form is equally induced by S23906 in wt and BRCA2-deficient cells, likely indicating a role for BRCA2 downstream of Chk1. Accordingly, override of the S phase arrest by either 7-hydroxystaurosporine (UCN-01) or AZD7762 potentiates the cytotoxic activity of S23906 in wt, but not in BRCA2-deficient cells. Together, our findings suggest that the pronounced sensitivity of BRCA2-deficient cells to S23906 is due to both a defective S-phase arrest and the absence of HR repair. Tumors with deficiencies for proteins involved in HR, and BRCA2 in particular, may thus show increased sensitivity to S23906, thereby providing a rationale for patient selection in clinical trials.
Collapse
Key Words
- ATR, Ataxia telangiectasia- and RAD3-related
- DNA alkylators
- DNA double strand breaks
- DNA replication
- DSBs, Double Strand Breaks
- FA, Fanconi Anemia
- GAPDH, Glyceraldehyde-3-phosphate dehydrogenase
- HR, Homologous Recombination
- HU, Hydroxyurea
- Homologous recombination
- ICLs, Inter-strand Crosslinks
- NER, Nucleotide Excision Repair
- NHEJ, Non-Homologous End-Joining
- TCR, Transcription-Coupled Repair
- UCN-01, 7-hydroxystaurosporine.
- checkpoint control
Collapse
Affiliation(s)
- Céline J Rocca
- a Laboratory of Cancer Biology and Therapeutics ; Centre de Recherche Saint-Antoine ; Paris , France
| | | | | | | | | | | |
Collapse
|
27
|
Al-Gholam MA, Nooh HZ, El-Mehi AE, El-Barbary AEM, Fokar AZE. Protective effect of rosemary on acrylamide motor neurotoxicity in spinal cord of rat offspring: postnatal follow-up study. Anat Cell Biol 2016; 49:34-49. [PMID: 27051566 PMCID: PMC4819076 DOI: 10.5115/acb.2016.49.1.34] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 12/17/2022] Open
Abstract
The direct interactive effects of rosemary and acrylamide on the development of motor neurons in the spinal cord remains unknown. Our goal is to confirm the protective effects of rosemary against motor neuronal degeneration induced by acrylamide in the developing postnatal rat spinal cord using a postnatal rat model. We assigned the offspring of treated female rats into control, rosemary; acrylamide group; and recovery groups. This work depended on clinical, histopathological, morphometrically, immunohistochemical and genetic methods. In the acrylamide group, we observed oxidation, motor neuron degeneration, apoptosis, myelin degeneration, neurofilament reduction, reactive gliosis. Whoever, concomitant rosemary intake and withdrawal of acrylamide modulate these effects. These findings proof that dietary rosemary can directly protect motor neuron against acrylamide toxicity in the mammalian developing spinal cord.
Collapse
Affiliation(s)
- Marwa A Al-Gholam
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Hanaa Zakaria Nooh
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Abeer E El-Mehi
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Abd El-Moneum El-Barbary
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Ahmed Zo El Fokar
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| |
Collapse
|
28
|
Jacobs K, Zambelli F, Mertzanidou A, Smolders I, Geens M, Nguyen HT, Barbé L, Sermon K, Spits C. Higher-Density Culture in Human Embryonic Stem Cells Results in DNA Damage and Genome Instability. Stem Cell Reports 2016; 6:330-41. [PMID: 26923824 PMCID: PMC4788786 DOI: 10.1016/j.stemcr.2016.01.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/24/2016] [Accepted: 01/25/2016] [Indexed: 12/01/2022] Open
Abstract
Human embryonic stem cells (hESC) show great promise for clinical and research applications, but their well-known proneness to genomic instability hampers the development to their full potential. Here, we demonstrate that medium acidification linked to culture density is the main cause of DNA damage and genomic alterations in hESC grown on feeder layers, and this even in the short time span of a single passage. In line with this, we show that increasing the frequency of the medium refreshments minimizes the levels of DNA damage and genetic instability. Also, we show that cells cultured on laminin-521 do not present this increase in DNA damage when grown at high density, although the (long-term) impact on their genomic stability remains to be elucidated. Our results explain the high levels of genome instability observed over the years by many laboratories worldwide, and show that the development of optimal culture conditions is key to solving this problem. Increased culture density induces DNA damage and genomic alterations in hESC Medium acidification due to lactic acid accumulation is the main driver More frequent medium refreshments rescues genomic integrity in high-density culture Laminin-521 reduces DNA damage but has no clear effect on genomic instability
Collapse
Affiliation(s)
- Kurt Jacobs
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; Institute of Molecular Cancer Research, University of Zurich (UZH), Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Filippo Zambelli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Afroditi Mertzanidou
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ilse Smolders
- Research Group Experimental Neuropharmacology, Center for Neurosciences C4N, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Mieke Geens
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ha Thi Nguyen
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium; Center for Molecular Biology, Institute of Research and Development, Duy Tan University, K7/25 Quang Trung, Danang 550000, Vietnam
| | - Lise Barbé
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Karen Sermon
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Claudia Spits
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
29
|
Petroni M, Sardina F, Heil C, Sahún-Roncero M, Colicchia V, Veschi V, Albini S, Fruci D, Ricci B, Soriani A, Di Marcotullio L, Screpanti I, Gulino A, Giannini G. The MRN complex is transcriptionally regulated by MYCN during neural cell proliferation to control replication stress. Cell Death Differ 2016; 23:197-206. [PMID: 26068589 PMCID: PMC4716299 DOI: 10.1038/cdd.2015.81] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 04/12/2015] [Accepted: 05/18/2015] [Indexed: 12/27/2022] Open
Abstract
The MRE11/RAD50/NBS1 (MRN) complex is a major sensor of DNA double strand breaks, whose role in controlling faithful DNA replication and preventing replication stress is also emerging. Inactivation of the MRN complex invariably leads to developmental and/or degenerative neuronal defects, the pathogenesis of which still remains poorly understood. In particular, NBS1 gene mutations are associated with microcephaly and strongly impaired cerebellar development, both in humans and in the mouse model. These phenotypes strikingly overlap those induced by inactivation of MYCN, an essential promoter of the expansion of neuronal stem and progenitor cells, suggesting that MYCN and the MRN complex might be connected on a unique pathway essential for the safe expansion of neuronal cells. Here, we show that MYCN transcriptionally controls the expression of each component of the MRN complex. By genetic and pharmacological inhibition of the MRN complex in a MYCN overexpression model and in the more physiological context of the Hedgehog-dependent expansion of primary cerebellar granule progenitor cells, we also show that the MRN complex is required for MYCN-dependent proliferation. Indeed, its inhibition resulted in DNA damage, activation of a DNA damage response, and cell death in a MYCN- and replication-dependent manner. Our data indicate the MRN complex is essential to restrain MYCN-induced replication stress during neural cell proliferation and support the hypothesis that replication-born DNA damage is responsible for the neuronal defects associated with MRN dysfunctions.
Collapse
Affiliation(s)
- M Petroni
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - F Sardina
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - C Heil
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - M Sahún-Roncero
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - V Colicchia
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - V Veschi
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - S Albini
- Paediatric Haematology/Oncology Department, IRCCS, Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy
| | - D Fruci
- Paediatric Haematology/Oncology Department, IRCCS, Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy
| | - B Ricci
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - A Soriani
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - L Di Marcotullio
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - I Screpanti
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - A Gulino
- Department Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - G Giannini
- Istituto Pasteur-Fondazione Cenci Bolognetti, Deptartment of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| |
Collapse
|
30
|
Poletto M, Legrand AJ, Fletcher SC, Dianov GL. p53 coordinates base excision repair to prevent genomic instability. Nucleic Acids Res 2016; 44:3165-75. [PMID: 26773055 PMCID: PMC4838360 DOI: 10.1093/nar/gkw015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/06/2016] [Indexed: 12/03/2022] Open
Abstract
DNA constantly undergoes chemical modification due to endogenous and exogenous mutagens. The DNA base excision repair (BER) pathway is the frontline mechanism handling the majority of these lesions, and primarily involves a DNA incision and subsequent resealing step. It is imperative that these processes are extremely well-coordinated as unrepaired DNA single strand breaks (SSBs) can be converted to DNA double strand breaks during replication thus triggering genomic instability. However, the mechanism(s) governing the BER process are poorly understood. Here we show that accumulation of unrepaired SSBs triggers a p53/Sp1-dependent downregulation of APE1, the endonuclease responsible for the DNA incision during BER. Importantly, we demonstrate that impaired p53 function, a characteristic of many cancers, leads to a failure of the BER coordination mechanism, overexpression of APE1, accumulation of DNA strand breaks and results in genomic instability. Our data provide evidence for a previously unrecognized mechanism for coordination of BER by p53, and its dysfunction in p53-inactivated cells.
Collapse
Affiliation(s)
- Mattia Poletto
- CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Department of Oncology, Old Road Campus Research Building, OX37DQ Oxford, UK
| | - Arnaud J Legrand
- CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Department of Oncology, Old Road Campus Research Building, OX37DQ Oxford, UK
| | - Sally C Fletcher
- CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Department of Oncology, Old Road Campus Research Building, OX37DQ Oxford, UK
| | - Grigory L Dianov
- CRUK & MRC Oxford Institute for Radiation Oncology, University of Oxford, Department of Oncology, Old Road Campus Research Building, OX37DQ Oxford, UK Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Lavrenteva 10, 630090 Novosibirsk, Russia
| |
Collapse
|
31
|
Costa PA, Poli JHZ, Sperotto NDM, Moura DJ, Saffi J, Nin MS, Barros HMT. Brain DNA damage and behavioral changes after repeated intermittent acute ethanol withdrawal by young rats. Psychopharmacology (Berl) 2015; 232:3623-36. [PMID: 26231496 DOI: 10.1007/s00213-015-4015-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 06/30/2015] [Indexed: 01/10/2023]
Abstract
RATIONALE Alcohol addiction causes severe problems, and its deprivation may potentiate symptoms such as anxiety. Furthermore, ethanol is a neurotoxic agent that induces degeneration and the consequences underlying alcohol-mediated brain damage remain unclear. OBJECTIVES This study assessed the behavioral changes during acute ethanol withdrawal periods and determined the levels of DNA damage and reactive oxygen species (ROS) in multiple brain areas. METHODS Male Wistar rats were subjected to an oral ethanol self-administration procedure with a forced diet where they were offered 8% (v/v) ethanol solution for 21 days followed by five repeated 24-h cycles alternating between ethanol withdrawal and re-exposure. Control animals received an isocaloric control diet without ethanol. Behavioral changes were analyzed on ethanol withdrawal days in the open-field (OF) and elevated plus-maze (EPM) tests within the first 6 h of ethanol deprivation. The pre-frontal cortex, hypothalamus, striatum, hippocampus, and cerebellum were dissected for alkaline and neutral comet assays and for dichlorofluorescein ROS testing. RESULTS The repeated intermittent ethanol access enhanced solution intake and alcohol-seeking behavior. Decreased exploratory activity was observed in the OF test, and the animals stretched less in the EPM test. DNA single-strand breaks and ROS production were significantly higher in all structures evaluated in the ethanol-treated rats compared with controls. CONCLUSIONS The animal model of repeated intermittent ethanol access induced behavioral changes in rats, and this ethanol exposure model induced an increase in DNA single-strand breaks and ROS production in all brain areas. Our results suggest that these brain damages may influence future behaviors.
Collapse
Affiliation(s)
- Priscila A Costa
- Laboratory of Neuropsycopharmacology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | | | | | | | | |
Collapse
|
32
|
Mirzayans R, Andrais B, Scott A, Wang YW, Weiss RH, Murray D. Spontaneous γH2AX Foci in Human Solid Tumor-Derived Cell Lines in Relation to p21WAF1 and WIP1 Expression. Int J Mol Sci 2015; 16:11609-28. [PMID: 26006237 PMCID: PMC4463719 DOI: 10.3390/ijms160511609] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/12/2015] [Accepted: 05/15/2015] [Indexed: 12/26/2022] Open
Abstract
Phosphorylation of H2AX on Ser139 (γH2AX) after exposure to ionizing radiation produces nuclear foci that are detectable by immunofluorescence microscopy. These so-called γH2AX foci have been adopted as quantitative markers for DNA double-strand breaks. High numbers of spontaneous γH2AX foci have also been reported for some human solid tumor-derived cell lines, but the molecular mechanism(s) for this response remains elusive. Here we show that cancer cells (e.g., HCT116; MCF7) that constitutively express detectable levels of p21WAF1 (p21) exhibit low numbers of γH2AX foci (<3/nucleus), whereas p21 knockout cells (HCT116p21−/−) and constitutively low p21-expressing cells (e.g., MDA-MB-231) exhibit high numbers of foci (e.g., >50/nucleus), and that these foci are not associated with apoptosis. The majority (>95%) of cells within HCT116p21−/− and MDA-MB-231 cultures contain high levels of phosphorylated p53, which is localized in the nucleus. We further show an inverse relationship between γH2AX foci and nuclear accumulation of WIP1, an oncogenic phosphatase. Our studies suggest that: (i) p21 deficiency might provide a selective pressure for the emergence of apoptosis-resistant progeny exhibiting genomic instability, manifested as spontaneous γH2AX foci coupled with phosphorylation and nuclear accumulation of p53; and (ii) p21 might contribute to positive regulation of WIP1, resulting in dephosphorylation of γH2AX.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| | - Bonnie Andrais
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| | - April Scott
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| | - Ying W Wang
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| | - Robert H Weiss
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, CA 95616, USA.
- Department of Medicine, Mather VA Medical Center, Sacramento, CA 95655, USA.
| | - David Murray
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
33
|
Renna C, Salaroli R, Cocchi C, Cenacchi G. XAV939-mediated ARTD activity inhibition in human MB cell lines. PLoS One 2015; 10:e0124149. [PMID: 25835728 PMCID: PMC4383513 DOI: 10.1371/journal.pone.0124149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 03/12/2015] [Indexed: 01/22/2023] Open
Abstract
Diphtheria toxin-like ADP-ribosyltransferases 1 and 5 (ARTD-1, ARTD-5) are poly ADP-ribose enzymes (PARP) involved in non-homologous end-joining (NHEJ), which is the major pathway of double-strand break (DSB) repair. In addition, ARTD-5, or Tankyrase (TNKS), is a positive regulator of the WNT signaling implicated in the development and biological behavior of many neoplasms, such as Medulloblastoma (MB), in which radiotherapy is an essential part of the treatment. The use of radiosensitizing agents may improve the therapeutic index in MB patients by increasing the efficacy of radiotherapy, while reducing toxicity to the neuroaxis. ARTD-5 seems to be a good molecular target for improving the current treatment of MB. In this study, we used the small molecule XAV939, a potent ARTD-5 inhibitor with a slight affinity for ARTD-1, in different human MB cell lines. XAV939 inhibited the WNT pathway and DNA-PKcs in our MB cells, with many biological consequences. The co-administration of XAV939 and ionizing radiations (IR) inhibited MB cells proliferation and clonogenic capacity, decreased their efficacy in repairing DNA damage, and increased IR-induced cell mortality. In conclusion, our in vitro data show that XAV939 could be a very promising small molecule in MB treatment, and these results lay the basis for further in vivo studies with the aim of improving the current therapy available for MB patients.
Collapse
Affiliation(s)
- Cristiano Renna
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Roberta Salaroli
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Claudia Cocchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
34
|
Kumar M, Kaur P, Kumar S, Kaur S. Antiproliferative and Apoptosis Inducing Effects of Non-Polar Fractions from Lawsonia inermis L. in Cervical (HeLa) Cancer Cells. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2015; 21:249-60. [PMID: 25931778 PMCID: PMC4411381 DOI: 10.1007/s12298-015-0285-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/13/2015] [Accepted: 03/02/2015] [Indexed: 05/03/2023]
Abstract
Two non-polar fractions viz. hexane (Hex-LI) and chloroform fraction (CHCl3-LI) of Lawsonia inermis were studied for their antiproliferative potential in various cancer cell lines viz. HeLa, MCF-7, A549 and C6 glioma cells. Both the fractions showed more than 60 % of growth inhibition in all the tested cell lines at highest tested concentration. In clonogenic assay, different concentrations of Hex-LI and CHCl3-LI decreased the number and size of colonies as compared to control in HeLa cells. The apoptotic effects as nuclear condensation, fragmentation were visualized with Hoechst-33342 staining of HeLa cells using confocal microscope. Both fractions induced apoptotic cell death in human cervical carcinoma (HeLa) cells as evident from flow cytometric analysis carried out using Annexin V-FITC and propidium iodide dyes. CHCl3-LI treated cells significantly induced apoptosis (25.43 %) in comparison to control. Results from Neutral Comet assay demonstrated that both fractions induced double stranded breaks (DSB's) in HeLa cells. Our data indicated that Hex-LI and CHCl3-LI treated cells showed significant increase of 32.2 and 18.56 % reactive oxygen species (ROS) levels in DCFH-DA assay respectively. Further, experimental studies to decipher exact pathway via which these fractions induce cell death are in progress.
Collapse
Affiliation(s)
- Manish Kumar
- />Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Paramjeet Kaur
- />Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Subodh Kumar
- />Department of Chemistry, UGC Centre for Advanced Studies, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Satwinderjeet Kaur
- />Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| |
Collapse
|
35
|
Khoronenkova SV, Dianov GL. ATM prevents DSB formation by coordinating SSB repair and cell cycle progression. Proc Natl Acad Sci U S A 2015; 112:3997-4002. [PMID: 25775545 PMCID: PMC4386361 DOI: 10.1073/pnas.1416031112] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
DNA single-strand breaks (SSBs) arise as a consequence of spontaneous DNA instability and are also formed as DNA repair intermediates. Their repair is critical because they otherwise terminate gene transcription and generate toxic DNA double-strand breaks (DSBs) on replication. To prevent the formation of DSBs, SSB repair must be completed before DNA replication. To accomplish this, cells should be able to detect unrepaired SSBs, and then delay cell cycle progression to allow more time for repair; however, to date there is no evidence supporting the coordination of SSB repair and replication in human cells. Here we report that ataxia-telangiectasia mutated kinase (ATM) plays a major role in restricting the replication of SSB-containing DNA and thus prevents DSB formation. We show that ATM is activated by SSBs and coordinates their repair with DNA replication. SSB-mediated ATM activation is followed by a G1 cell cycle delay that allows more time for repair and thus prevents the replication of damaged DNA and DSB accrual. These findings establish an unanticipated role for ATM in the signaling of DNA SSBs and provide important insight into the molecular defects leading to genetic instability in patients with ataxia-telangiectasia.
Collapse
Affiliation(s)
- Svetlana V Khoronenkova
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom; and Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Grigory L Dianov
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| |
Collapse
|
36
|
Vignardi CP, Hasue FM, Sartório PV, Cardoso CM, Machado ASD, Passos MJACR, Santos TCA, Nucci JM, Hewer TLR, Watanabe IS, Gomes V, Phan NV. Genotoxicity, potential cytotoxicity and cell uptake of titanium dioxide nanoparticles in the marine fish Trachinotus carolinus (Linnaeus, 1766). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 158:218-229. [PMID: 25481788 DOI: 10.1016/j.aquatox.2014.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 11/05/2014] [Accepted: 11/14/2014] [Indexed: 06/04/2023]
Abstract
Nanoparticles have physicochemical characteristics that make them useful in areas such as science, technology, medicine and in products of everyday use. Recently the manufacture and variety of these products has grown rapidly, raising concerns about their impact on human health and the environment. Adverse effects of exposure to nanoparticles have been reported for both terrestrial and aquatic organisms, but the toxic effects of the substances on marine organisms remain poorly understood. The main aim of this study was to evaluate the genotoxicity of TiO2-NP in the marine fish Trachinotus carolinus, through cytogenotoxic methods. The fish received two different doses of 1.5 μg and 3.0 μg-TiO2-NP g(-1) by intraperitoneal injection. Blood samples were collected to analyze erythrocyte viability using the Trypan Blue exclusion test, comet assay (pH>13), micronucleus (MN) and other erythrocyte nuclear abnormalities (ENA) 24, 48 and 72 h after injection. The possible cell uptake of TiO2-NP in fish injected with the higher dose was investigated after 72 h using transmission electron microscopy (TEM). The results showed that TiO2-NP is genotoxic and potentially cytotoxic for this species, causing DNA damage, inducing the formation of MN and other ENA, and decreasing erythrocyte viability. TEM examination revealed that cell uptake of TiO2-NP was mainly in the kidney, liver, gills and to a lesser degree in muscle. To the extent of the authors' knowledge, this is the first in vivo study of genotoxicity and other effects of TiO2-NP in a marine fish.
Collapse
Affiliation(s)
- Caroline P Vignardi
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Fabio M Hasue
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Priscila V Sartório
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Caroline M Cardoso
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Alex S D Machado
- Faculty of Veterinary Medicine, Integrated College North of Minas Osmane Barbosa Avenue, 11111, JK, Montes Claros, MG 39404006, Brazil.
| | - Maria J A C R Passos
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Thais C A Santos
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Juliana M Nucci
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Thiago L R Hewer
- Institute of Chemistry, University of São Paulo, Prof. Lineu Prestes Avenue, 748, Cidade Universitária, Butantã, São Paulo, SP 05508000, Brazil.
| | - Ii-Sei Watanabe
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Prof. Lineu Prestes Avenue, 2415, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Vicente Gomes
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| | - Ngan V Phan
- Department of Biological Oceanography, Oceanographic Institute, University of São Paulo, Praça do Oceanogáfico 191, Cidade Universitária, Butantã, São Paulo, SP 05508900, Brazil.
| |
Collapse
|
37
|
Ma Y, Chapman J, Levine M, Polireddy K, Drisko J, Chen Q. High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Sci Transl Med 2014; 6:222ra18. [PMID: 24500406 DOI: 10.1126/scitranslmed.3007154] [Citation(s) in RCA: 320] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ascorbate (vitamin C) was an early, unorthodox therapy for cancer, with an outstanding safety profile and anecdotal clinical benefit. Because oral ascorbate was ineffective in two cancer clinical trials, ascorbate was abandoned by conventional oncology but continued to be used in complementary and alternative medicine. Recent studies provide rationale for reexamining ascorbate treatment. Because of marked pharmacokinetic differences, intravenous, but not oral, ascorbate produces millimolar concentrations both in blood and in tissues, killing cancer cells without harming normal tissues. In the interstitial fluid surrounding tumor cells, millimolar concentrations of ascorbate exert local pro-oxidant effects by mediating hydrogen peroxide (H(2)O(2)) formation, which kills cancer cells. We investigated downstream mechanisms of ascorbate-induced cell death. Data show that millimolar ascorbate, acting as a pro-oxidant, induced DNA damage and depleted cellular adenosine triphosphate (ATP), activated the ataxia telangiectasia mutated (ATM)/adenosine monophosphate-activated protein kinase (AMPK) pathway, and resulted in mammalian target of rapamycin (mTOR) inhibition and death in ovarian cancer cells. The combination of parenteral ascorbate with the conventional chemotherapeutic agents carboplatin and paclitaxel synergistically inhibited ovarian cancer in mouse models and reduced chemotherapy-associated toxicity in patients with ovarian cancer. On the basis of its potential benefit and minimal toxicity, examination of intravenous ascorbate in combination with standard chemotherapy is justified in larger clinical trials.
Collapse
Affiliation(s)
- Yan Ma
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | | | | | |
Collapse
|
38
|
Sutherland JM, Fraser BA, Sobinoff AP, Pye VJ, Davidson TL, Siddall NA, Koopman P, Hime GR, McLaughlin EA. Developmental Expression of Musashi-1 and Musashi-2 RNA-Binding Proteins During Spermatogenesis: Analysis of the Deleterious Effects of Dysregulated Expression1. Biol Reprod 2014; 90:92. [DOI: 10.1095/biolreprod.113.115261] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
39
|
|
40
|
Ciszewski WM, Wagner W, Kania KD, Dastych J. Interleukin-4 enhances PARP-dependent DNA repair activity in vitro. J Interferon Cytokine Res 2014; 34:734-40. [PMID: 24724620 DOI: 10.1089/jir.2014.0029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Eukaryotic cells possess several DNA repair mechanisms, including homologous recombination and the non-homologous end-joining (NHEJ) system. There are two known NHEJ systems. The major mechanism depends on the catalytic unit of DNA-dependent protein kinase (DNA-PKcs) and DNA ligase IV, and an alternative mechanism (B-NHEJ) depends on poly(ADP-ribose) polymerase (PARP). These systems are upregulated by genotoxic agents. Interleukin 4 (IL-4) is an immunoregulatory cytokine that is secreted by immune cells upon contact with certain genotoxic compounds and is known to regulate several genes encoding components of DNA repair systems in human monocytes. We have investigated the possible effects of IL-4 on the DNA repair process within murine and human cells exposed to selected genotoxic compounds. In a series of experiments, including the comet assay, cell surface annexin V staining, analysis of histone H2AX phosphorylation, and a DNA end-joining assay, we observed that IL-4 decreased DNA damage in murine fibroblasts and human glioblastoma cells exposed to genotoxic agents and increased DNA ligation activity in the nuclei of these cells in a process that depended on PARP. These observations suggest that IL-4 is capable of upregulating the alternative NHEJ DNA repair mechanism in murine and human cells.
Collapse
Affiliation(s)
- Wojciech Michał Ciszewski
- 1 Laboratory of Cellular Immunology, Institute of Medical Biology , Polish Academy of Sciences, Lodz, Poland
| | | | | | | |
Collapse
|
41
|
Clynes D, Jelinska C, Xella B, Ayyub H, Taylor S, Mitson M, Bachrati CZ, Higgs DR, Gibbons RJ. ATRX dysfunction induces replication defects in primary mouse cells. PLoS One 2014; 9:e92915. [PMID: 24651726 PMCID: PMC3961441 DOI: 10.1371/journal.pone.0092915] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/27/2014] [Indexed: 01/09/2023] Open
Abstract
The chromatin remodeling protein ATRX, which targets tandem repetitive DNA, has been shown to be required for expression of the alpha globin genes, for proliferation of a variety of cellular progenitors, for chromosome congression and for the maintenance of telomeres. Mutations in ATRX have recently been identified in tumours which maintain their telomeres by a telomerase independent pathway involving homologous recombination thought to be triggered by DNA damage. It is as yet unknown whether there is a central underlying mechanism associated with ATRX dysfunction which can explain the numerous cellular phenomena observed. There is, however, growing evidence for its role in the replication of various repetitive DNA templates which are thought to have a propensity to form secondary structures. Using a mouse knockout model we demonstrate that ATRX plays a direct role in facilitating DNA replication. Ablation of ATRX alone, although leading to a DNA damage response at telomeres, is not sufficient to trigger the alternative lengthening of telomere pathway in mouse embryonic stem cells.
Collapse
Affiliation(s)
- David Clynes
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Clare Jelinska
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Barbara Xella
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Helena Ayyub
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Stephen Taylor
- Computational Biology Research Group, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Matthew Mitson
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Csanád Z Bachrati
- School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, United Kingdom
| | - Douglas R Higgs
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Richard J Gibbons
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
42
|
Lee MH, Wang L, Chang ZF. The contribution of mitochondrial thymidylate synthesis in preventing the nuclear genome stress. Nucleic Acids Res 2014; 42:4972-84. [PMID: 24561807 PMCID: PMC4005647 DOI: 10.1093/nar/gku152] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In quiescent fibroblasts, the expression levels of cytosolic enzymes for thymidine triphosphate (dTTP) synthesis are down-regulated, causing a marked reduction in the dTTP pool. In this study, we provide evidence that mitochondrial thymidylate synthesis via thymidine kinase 2 (TK2) is a limiting factor for the repair of ultraviolet (UV) damage in the nuclear compartment in quiescent fibroblasts. We found that TK2 deficiency causes secondary DNA double-strand breaks formation in the nuclear genome of quiescent cells at the late stage of recovery from UV damage. Despite slower repair of quiescent fibroblast deficient in TK2, DNA damage signals eventually disappeared, and these cells were capable of re-entering the S phase after serum stimulation. However, these cells displayed severe genome stress as revealed by the dramatic increase in 53BP1 nuclear body in the G1 phase of the successive cell cycle. Here, we conclude that mitochondrial thymidylate synthesis via TK2 plays a role in facilitating the quality repair of UV damage for the maintenance of genome integrity in the cells that are temporarily arrested in the quiescent state.
Collapse
Affiliation(s)
- Ming-Hsiang Lee
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan (R.O.C.), Department of Anatomy, Physiology and Biochemistry, The Swedish University of Agricultural Sciences, Biomedical Center, S-751 23 Uppsala, Sweden and Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan (R.O.C.)
| | | | | |
Collapse
|
43
|
Abstract
Thioredoxin-1 (Trx1) is an antioxidant enzyme with a protective role in the removal of oxidative stress. We investigated the mechanism by which the redox modulator Trx1 affects base excision repair (BER) activity to understand the protective role of Trx1. We constructed a Trx1 knockdown system to demonstrate the specific mechanism of Trx1 shRNA cells compared with that in the wild type cells, leading to increased cellular susceptibility to a sublethal dose of BER-inducible toxicant, nitrosomethylurea (NMU). In addition, we observed a modulatory role of Trx1 in the BER pathway via the p53 downstream gene, growth arrest, and DNA-damage-inducible protein 45 α (Gadd45a). The protein level and function of p53, a Trx1 downstream gene, coincidently decreased in the Trx1 shRNA cells. Futhermore, Trx1 shRNA cells showed decreased Gadd45a expression and interaction of Gadd45a with apurinic/apyrimidinic endonuclease 1 (APE1) as well as APE activity. In conclusion, Trx1 might cooperate in the control of APE1 function by modulating the p53-mediated BER via the protein-protein interaction between Gadd45a and APE1, providing insight into the novel role of redox factor Trx1 in modulation of BER.
Collapse
|
44
|
Bhosle J, Kiakos K, Porter ACG, Wu J, Makris A, Hartley JA, Hochhauser D. Treatment with gefitinib or lapatinib induces drug resistance through downregulation of topoisomerase IIα expression. Mol Cancer Ther 2013; 12:2897-908. [PMID: 24092808 DOI: 10.1158/1535-7163.mct-12-1049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The EGF receptor (EGFR) is therapeutically targeted by antibodies and small molecules in solid tumors including lung, colorectal, and breast cancer. However, chemotherapy remains important, and efforts to improve efficacy through combination with targeted agents is challenging. This study examined the effects of short and long durations of exposure to the EGFR- and HER2-targeted tyrosine kinase inhibitors (TKI) gefitinib and lapatinib, on induction of cell death and DNA damage by topoisomerase IIα (Topo IIα) poisons, in the SK-Br-3 HER2-amplified breast cancer cell line. Short exposure to either gefitinib or lapatinib for 1 hour did not affect the induction of apoptosis by the Topo IIα poisons doxorubicin, etoposide, and m-AMSA. In contrast, cells treated for 48 hours were resistant to all three drugs. Short exposure (1 hour) to TKI did not alter the number of DNA single- or double-strand breaks (DSB) induced, whereas longer exposure (48 hours) reduced the number of DNA DSBs and the formation of γ-H2AX foci. Both gefitinib and lapatinib reduced the expression and activity of Topo IIα at 48 hours. Studies using a cell line with inducible downregulation of Topo IIα showed that expression of Topo IIα, and not Topo IIβ, determined the number of DNA strand breaks induced by these chemotherapeutic agents. These results indicate that prolonged exposure to TKIs targeting EGFR and HER2 induce resistance to doxorubicin, etoposide, and m-AMSA through downregulation of Topo IIα. This may explain why their addition to chemotherapy regimens have not increased efficacy.
Collapse
Affiliation(s)
- Jaishree Bhosle
- Corresponding Author: Daniel Hochhauser, UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London WC1E 6BT, UK.
| | | | | | | | | | | | | |
Collapse
|
45
|
Milić M, Kišan M, Rogulj D, Radman M, Lovrenčić MV, Konjevoda P, Domijan AM. Level of primary DNA damage in the early stage of metabolic syndrome. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2013; 758:1-5. [DOI: 10.1016/j.mrgentox.2013.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 06/27/2013] [Accepted: 07/05/2013] [Indexed: 11/15/2022]
|
46
|
HDAC inhibitor confers radiosensitivity to prostate stem-like cells. Br J Cancer 2013; 109:3023-33. [PMID: 24220693 PMCID: PMC3859953 DOI: 10.1038/bjc.2013.691] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/05/2013] [Accepted: 10/09/2013] [Indexed: 01/06/2023] Open
Abstract
Background: Radiotherapy can be an effective treatment for prostate cancer, but radiorecurrent tumours do develop. Considering prostate cancer heterogeneity, we hypothesised that primitive stem-like cells may constitute the radiation-resistant fraction. Methods: Primary cultures were derived from patients undergoing resection for prostate cancer or benign prostatic hyperplasia. After short-term culture, three populations of cells were sorted, reflecting the prostate epithelial hierarchy, namely stem-like cells (SCs, α2β1integrinhi/CD133+), transit-amplifying (TA, α2β1integrinhi/CD133−) and committed basal (CB, α2β1integrinlo) cells. Radiosensitivity was measured by colony-forming efficiency (CFE) and DNA damage by comet assay and DNA damage foci quantification. Immunofluorescence and flow cytometry were used to measure heterochromatin. The HDAC (histone deacetylase) inhibitor Trichostatin A was used as a radiosensitiser. Results: Stem-like cells had increased CFE post irradiation compared with the more differentiated cells (TA and CB). The SC population sustained fewer lethal double-strand breaks than either TA or CB cells, which correlated with SCs being less proliferative and having increased levels of heterochromatin. Finally, treatment with an HDAC inhibitor sensitised the SCs to radiation. Interpretation: Prostate SCs are more radioresistant than more differentiated cell populations. We suggest that the primitive cells survive radiation therapy and that pre-treatment with HDAC inhibitors may sensitise this resistant fraction.
Collapse
|
47
|
Shee C, Cox BD, Gu F, Luengas EM, Joshi MC, Chiu LY, Magnan D, Halliday JA, Frisch RL, Gibson JL, Nehring RB, Do HG, Hernandez M, Li L, Herman C, Hastings PJ, Bates D, Harris RS, Miller KM, Rosenberg SM. Engineered proteins detect spontaneous DNA breakage in human and bacterial cells. eLife 2013; 2:e01222. [PMID: 24171103 PMCID: PMC3809393 DOI: 10.7554/elife.01222] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/16/2013] [Indexed: 01/10/2023] Open
Abstract
Spontaneous DNA breaks instigate genomic changes that fuel cancer and evolution, yet direct quantification of double-strand breaks (DSBs) has been limited. Predominant sources of spontaneous DSBs remain elusive. We report synthetic technology for quantifying DSBs using fluorescent-protein fusions of double-strand DNA end-binding protein, Gam of bacteriophage Mu. In Escherichia coli GamGFP forms foci at chromosomal DSBs and pinpoints their subgenomic locations. Spontaneous DSBs occur mostly one per cell, and correspond with generations, supporting replicative models for spontaneous breakage, and providing the first true breakage rates. In mammalian cells GamGFP-labels laser-induced DSBs antagonized by end-binding protein Ku; co-localizes incompletely with DSB marker 53BP1 suggesting superior DSB-specificity; blocks resection; and demonstrates DNA breakage via APOBEC3A cytosine deaminase. We demonstrate directly that some spontaneous DSBs occur outside of S phase. The data illuminate spontaneous DNA breakage in E. coli and human cells and illustrate the versatility of fluorescent-Gam for interrogation of DSBs in living cells. DOI:http://dx.doi.org/10.7554/eLife.01222.001.
Collapse
Affiliation(s)
- Chandan Shee
- Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , United States ; Department of Molecular Virology and Microbiology , Baylor College of Medicine , Houston , United States ; Dan L Duncan Cancer Center, Baylor College of Medicine , Houston , United States ; Department of Biochemistry, Molecular Biology , Baylor College of Medicine , Houston , United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
da Silva ALG, da Rosa HT, Karnopp TE, Charlier CF, Ellwanger JH, Moura DJ, Possuelo LG, Valim ARDM, Guecheva TN, Henriques JAP. Evaluation of DNA damage in COPD patients and its correlation with polymorphisms in repair genes. BMC MEDICAL GENETICS 2013; 14:93. [PMID: 24053728 PMCID: PMC3848611 DOI: 10.1186/1471-2350-14-93] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/11/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND We investigated a potential link between genetic polymorphisms in genes XRCC1 (Arg399Gln), OGG1 (Ser326Cys), XRCC3 (Thr241Met), and XRCC4 (Ile401Thr) with the level of DNA damage and repair, accessed by comet and micronucleus test, in 51 COPD patients and 51 controls. METHODS Peripheral blood was used to perform the alkaline and neutral comet assay; and genetic polymorphisms by PCR/RFLP. To assess the susceptibility to exogenous DNA damage, the cells were treated with methyl methanesulphonate for 1-h or 3-h. After 3-h treatment the % residual damage was calculated assuming the value of 1-h treatment as 100%. The cytogenetic damage was evaluated by buccal micronucleus cytome assay (BMCyt). RESULTS COPD patients with the risk allele XRCC1 (Arg399Gln) and XRCC3 (Thr241Met) showed higher DNA damage by comet assay. The residual damage was higher for COPD with risk allele in the four genes. In COPD patients was showed negative correlation between BMCyt (binucleated, nuclear bud, condensed chromatin and karyorrhexic cells) with pulmonary function and some variant genotypes. CONCLUSION Our results suggest a possible association between variant genotypes in XRCC1 (Arg399Gln), OGG1 (Ser326Cys), XRCC3 (Thr241Met), and XRCC4 (Ile401Thr), DNA damage and progression of COPD.
Collapse
Affiliation(s)
- Andréa Lúcia Gonçalves da Silva
- Santa Cruz Hospital and Department of Health and Physical Education, University of Santa Cruz do Sul - UNISC, Avenida Independência, 2293, Bloco 42, Bairro Universitário, Santa Cruz do Sul, RS, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Changes in the number of double-strand DNA breaks in Chinese hamster V79 cells exposed to γ-radiation with different dose rates. Int J Mol Sci 2013; 14:13719-26. [PMID: 23880845 PMCID: PMC3742213 DOI: 10.3390/ijms140713719] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 06/15/2013] [Accepted: 06/19/2013] [Indexed: 12/12/2022] Open
Abstract
A comparative investigation of the induction of double-strand DNA breaks (DSBs) in the Chinese hamster V79 cells by γ-radiation at dose rates of 1, 10 and 400 mGy/min (doses ranged from 0.36 to 4.32 Gy) was performed. The acute radiation exposure at a dose rate of 400 mGy/min resulted in the linear dose-dependent increase of the γ-H2AX foci formation. The dose-response curve for the acute exposure was well described by a linear function y = 1.22 + 19.7x, where "y" is an average number of γ-H2AX foci per a cell and "x" is the absorbed dose (Gy). The dose rate reduction down to 10 mGy/min lead to a decreased number of γ-H2AX foci, as well as to a change of the dose-response relationship. Thus, the foci number up to 1.44 Gy increased and reached the "plateau" area between 1.44 and 4.32 Gy. There was only a slight increase of the γ-H2AX foci number (up to 7) in cells after the protracted exposure (up to 72 h) to ionizing radiation at a dose rate of 1 mGy/min. Similar effects of the varying dose rates were obtained when DNA damage was assessed using the comet assay. In general, our results show that the reduction of the radiation dose rate resulted in a significant decrease of DSBs per cell per an absorbed dose.
Collapse
|
50
|
Shin SW, Jung E, Kim S, Kim JH, Kim EG, Lee J, Park D. Antagonizing effects and mechanisms of afzelin against UVB-induced cell damage. PLoS One 2013; 8:e61971. [PMID: 23626759 PMCID: PMC3633960 DOI: 10.1371/journal.pone.0061971] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 03/15/2013] [Indexed: 11/19/2022] Open
Abstract
Ultraviolet (UV) radiation induces DNA damage, oxidative stress, and inflammatory processes in human keratinocytes, resulting in skin inflammation, photoaging, and photocarcinogenesis. Adequate protection of skin against the harmful effects of UV irradiation is essential. Therefore, in this study, we investigated the protective effects of afzelin, one of the flavonoids, against UV irradiation in human keratinocytes and epidermal equivalent models. Spectrophotometric measurements revealed that the afzelin extinction maxima were in the UVB and UVA range, and UV transmission below 376 nm was <10%, indicating UV-absorbing activity of afzelin. In the phototoxicity assay using the 3T3 NRU phototoxicity test (3T3-NRU-PT), afzelin presented a tendency to no phototoxic potential. In addition, in order to investigate cellular functions of afzelin itself, cells were treated with afzelin after UVB irradiation. In human keratinocyte, afzelin effectively inhibited the UVB-mediated increase in lipid peroxidation and the formation of cyclobutane pyrimidine dimers. Afzelin also inhibited UVB-induced cell death in human keratinocytes by inhibiting intrinsic apoptotic signaling. Furthermore, afzelin showed inhibitory effects on UVB-induced release of pro-inflammatory mediators such as interleukin-6, tumor necrosis factor-α, and prostaglandin-E2 in human keratinocytes by interfering with the p38 kinase pathway. Using an epidermal equivalent model exposed to UVB radiation, anti-apoptotic activity of afzelin was also confirmed together with a photoprotective effect at the morphological level. Taken together, our results suggest that afzelin has several cellular activities such as DNA-protective, antioxidant, and anti-inflammatory as well as UV-absorbing activity and may protect human skin from UVB-induced damage by a combination of UV-absorbing and cellular activities.
Collapse
Affiliation(s)
- Seoung Woo Shin
- Biospectrum Life Science Institute, Seoungnam City, Gyunggi Do, Korea
| | - Eunsun Jung
- Biospectrum Life Science Institute, Seoungnam City, Gyunggi Do, Korea
| | - Seungbeom Kim
- Biospectrum Life Science Institute, Seoungnam City, Gyunggi Do, Korea
| | - Jang-Hyun Kim
- Dermiskin Life Science Institute, Pyeongtaek City, Gyunggi Do, Korea
| | - Eui-Gyun Kim
- ChiroChem Co., Ltd. Hannam University Science Park, Daejeon, Korea
| | - Jongsung Lee
- Department of Dermatological Health Management, Eulji University, Seongnam, Korea
- * E-mail: (JL); (DP)
| | - Deokhoon Park
- Biospectrum Life Science Institute, Seoungnam City, Gyunggi Do, Korea
- * E-mail: (JL); (DP)
| |
Collapse
|