101
|
Chen J, Huo J, Jia Z, Song Y, Li Y, Zhang L. Effects of atrazine on the proliferation and cytotoxicity of murine lymphocytes with the use of carboxyfluorescein succinimidyl ester-based flow cytometric approaches. Food Chem Toxicol 2015; 76:61-9. [DOI: 10.1016/j.fct.2014.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 10/24/2014] [Accepted: 11/28/2014] [Indexed: 12/31/2022]
|
102
|
Merkes C, Turkalo TK, Wilder N, Park H, Wenger LW, Lewin SJ, Azuma M. Ewing sarcoma ewsa protein regulates chondrogenesis of Meckel's cartilage through modulation of Sox9 in zebrafish. PLoS One 2015; 10:e0116627. [PMID: 25617839 PMCID: PMC4305327 DOI: 10.1371/journal.pone.0116627] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/11/2014] [Indexed: 11/19/2022] Open
Abstract
Ewing sarcoma is the second most common skeletal (bone and cartilage) cancer in adolescents, and it is characterized by the expression of the aberrant chimeric fusion gene EWS/FLI1. Wild-type EWS has been proposed to play a role in mitosis, splicing and transcription. We have previously shown that EWS/FLI1 interacts with EWS, and it inhibits EWS activity in a dominant manner. Ewing sarcoma is a cancer that specifically develops in skeletal tissues, and although the above data suggests the significance of EWS, its role in chondrogenesis/skeletogenesis is not understood. To elucidate the function of EWS in skeletal development, we generated and analyzed a maternal zygotic (MZ) ewsa/ewsa line because the ewsa/wt and ewsa/ewsa zebrafish appeared to be normal and fertile. Compared with wt/wt, the Meckel's cartilage of MZ ewsa/ewsa mutants had a higher number of craniofacial prehypertrophic chondrocytes that failed to mature into hypertrophic chondrocytes at 4 days post-fertilization (dpf). Ewsa interacted with Sox9, which is the master transcription factor for chondrogenesis. Sox9 target genes were either upregulated (ctgfa, ctgfb, col2a1a, and col2a1b) or downregulated (sox5, nog1, nog2, and bmp4) in MZ ewsa/ewsa embryos compared with the wt/wt zebrafish embryos. Among these Sox9 target genes, the chromatin immunoprecipitation (ChIP) experiment demonstrated that Ewsa directly binds to ctgfa and ctgfb loci. Consistently, immunohistochemistry showed that the Ctgf protein is upregulated in the Meckel's cartilage of MZ ewsa/ewsa mutants. Together, we propose that Ewsa promotes the differentiation from prehypertrophic chondrocytes to hypertrophic chondrocytes of Meckel's cartilage through inhibiting Sox9 binding site of the ctgf gene promoter. Because Ewing sarcoma specifically develops in skeletal tissue that is originating from chondrocytes, this new role of EWS may provide a potential molecular basis of its pathogenesis.
Collapse
Affiliation(s)
- Chris Merkes
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
| | - Timothy K. Turkalo
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
| | - Nicole Wilder
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
| | - Hyewon Park
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
| | - Luke W. Wenger
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
| | - Seth J. Lewin
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
| | - Mizuki Azuma
- Molecular Biosciences, University of Kansas, 7031 Haworth, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States of America
- * E-mail:
| |
Collapse
|
103
|
Hayano T, Yokota Y, Hosomichi K, Nakaoka H, Yoshihara K, Adachi S, Kashima K, Tsuda H, Moriya T, Tanaka K, Enomoto T, Inoue I. Molecular characterization of an intact p53 pathway subtype in high-grade serous ovarian cancer. PLoS One 2014; 9:e114491. [PMID: 25460179 PMCID: PMC4252108 DOI: 10.1371/journal.pone.0114491] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 11/10/2014] [Indexed: 12/30/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the most aggressive histological type of epithelial ovarian cancer, which is characterized by a high frequency of somatic TP53 mutations. We performed exome analyses of tumors and matched normal tissues of 34 Japanese patients with HGSOC and observed a substantial number of patients without TP53 mutation (24%, 8/34). Combined with the results of copy number variation analyses, we subdivided the 34 patients with HGSOC into subtypes designated ST1 and ST2. ST1 showed intact p53 pathway and was characterized by fewer somatic mutations and copy number alterations. In contrast, the p53 pathway was impaired in ST2, which is characterized by abundant somatic mutations and copy number alterations. Gene expression profiles combined with analyses using the Gene Ontology resource indicate the involvement of specific biological processes (mitosis and DNA helicase) that are relevant to genomic stability and cancer etiology. In particular we demonstrate the presence of a novel subtype of patients with HGSOC that is characterized by an intact p53 pathway, with limited genomic alterations and specific gene expression profiles.
Collapse
Affiliation(s)
- Takahide Hayano
- Division of Human Genetics, National Institute of Genetics, Mishima, Japan
| | - Yuki Yokota
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Hirofumi Nakaoka
- Division of Human Genetics, National Institute of Genetics, Mishima, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sosuke Adachi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Katsunori Kashima
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, Tokorozawa, Japan
| | - Takuya Moriya
- Department of Pathology, Kawasaki Medical School, Kurashiki, Japan
| | - Kenichi Tanaka
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Niigata Medical Center Hospital, Niigata, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ituro Inoue
- Division of Human Genetics, National Institute of Genetics, Mishima, Japan
| |
Collapse
|
104
|
Werner E, Wang H, Doetsch PW. Opposite roles for p38MAPK-driven responses and reactive oxygen species in the persistence and resolution of radiation-induced genomic instability. PLoS One 2014; 9:e108234. [PMID: 25271419 PMCID: PMC4182705 DOI: 10.1371/journal.pone.0108234] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 08/27/2014] [Indexed: 01/26/2023] Open
Abstract
We report the functional and temporal relationship between cellular phenotypes such as oxidative stress, p38MAPK-dependent responses and genomic instability persisting in the progeny of cells exposed to sparsely ionizing low-Linear Energy Transfer (LET) radiation such as X-rays or high-charge and high-energy (HZE) particle high-LET radiation such as 56Fe ions. We found that exposure to low and high-LET radiation increased reactive oxygen species (ROS) levels as a threshold-like response induced independently of radiation quality and dose. This response was sustained for two weeks, which is the period of time when genomic instability is evidenced by increased micronucleus formation frequency and DNA damage associated foci. Indicators for another persisting response sharing phenotypes with stress-induced senescence, including beta galactosidase induction, increased nuclear size, p38MAPK activation and IL-8 production, were induced in the absence of cell proliferation arrest during the first, but not the second week following exposure to high-LET radiation. This response was driven by a p38MAPK-dependent mechanism and was affected by radiation quality and dose. This stress response and elevation of ROS affected genomic instability by distinct pathways. Through interference with p38MAPK activity, we show that radiation-induced stress phenotypes promote genomic instability. In contrast, exposure to physiologically relevant doses of hydrogen peroxide or increasing endogenous ROS levels with a catalase inhibitor reduced the level of genomic instability. Our results implicate persistently elevated ROS following exposure to radiation as a factor contributing to genome stabilization.
Collapse
Affiliation(s)
- Erica Werner
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (PWD); (EW)
| | - Huichen Wang
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Paul W. Doetsch
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Hematology and Medical Oncology Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (PWD); (EW)
| |
Collapse
|
105
|
Baldini E, Tuccilli C, Prinzi N, Sorrenti S, Antonelli A, Gnessi L, Morrone S, Moretti C, Bononi M, Arlot-Bonnemains Y, D'Armiento M, Ulisse S. Effects of selective inhibitors of Aurora kinases on anaplastic thyroid carcinoma cell lines. Endocr Relat Cancer 2014; 21:797-811. [PMID: 25074669 DOI: 10.1530/erc-14-0299] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Aurora kinases are serine/threonine kinases that play an essential role in cell division. Their aberrant expression and/or function induce severe mitotic abnormalities, resulting in either cell death or aneuploidy. Overexpression of Aurora kinases is often found in several malignancies, among which is anaplastic thyroid carcinoma (ATC). We have previously demonstrated the in vitro efficacy of Aurora kinase inhibitors in restraining cell growth and survival of different ATC cell lines. In this study, we sought to establish which Aurora might represent the preferential drug target for ATC. To this end, the effects of two selective inhibitors of Aurora-A (MLN8237) and Aurora-B (AZD1152) on four human ATC cell lines (CAL-62, BHT-101, 8305C, and 8505C) were analysed. Both inhibitors reduced cell proliferation in a time- and dose-dependent manner, with IC50 ranges of 44.3-134.2 nM for MLN8237 and of 9.2-461.3 nM for AZD1152. Immunofluorescence experiments and time-lapse videomicroscopy yielded evidence that each inhibitor induced distinct mitotic phenotypes, but both of them prevented the completion of cytokinesis. As a result, poliploidy increased in all AZD1152-treated cells, and in two out of four cell lines treated with MLN8237. Apoptosis was induced in all the cells by MLN8237, and in BHT-101, 8305C, and 8505C by AZD1152, while CAL-62 exposed to AZD1152 died through necrosis after multiple rounds of endoreplication. Both inhibitors were capable of blocking anchorage-independent cell growth. In conclusion, we demonstrated that either Aurora-A or Aurora-B might represent therapeutic targets for the ATC treatment, but inhibition of Aurora-A appears more effective for suppressing ATC cell proliferation and for inducing the apoptotic pathway.
Collapse
Affiliation(s)
- Enke Baldini
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Chiara Tuccilli
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Natalie Prinzi
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Salvatore Sorrenti
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Alessandro Antonelli
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Lucio Gnessi
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Stefania Morrone
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Costanzo Moretti
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Marco Bononi
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Yannick Arlot-Bonnemains
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Massimino D'Armiento
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| | - Salvatore Ulisse
- Departments of Experimental MedicineSurgical Sciences'Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyDepartment of Internal MedicineUniversity of Pisa, Pisa, ItalyDepartment of Systems' MedicineUniversity of Rome Tor Vergata, Rome, ItalyDepartment of Surgery 'Pietro Valdoni''Sapienza' University of Rome, Viale Regina Elena, 324, 00161 Rome, ItalyCNRS - UMR 6290 (IGDR)University of Rennes 1, Rennes, France
| |
Collapse
|
106
|
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
|
107
|
Acentrosomal Drosophila epithelial cells exhibit abnormal cell division, leading to cell death and compensatory proliferation. Dev Cell 2014; 30:731-45. [PMID: 25241934 DOI: 10.1016/j.devcel.2014.08.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/01/2014] [Accepted: 08/07/2014] [Indexed: 12/14/2022]
Abstract
Mitotic spindles are critical for accurate chromosome segregation. Centrosomes, the primary microtubule nucleating centers of animal cells, play key roles in forming and orienting mitotic spindles. However, the survival of Drosophila without centrosomes suggested they are dispensable in somatic cells, challenging the canonical view. We used fly wing disc epithelia as a model to resolve these conflicting hypotheses, revealing that centrosomes play vital roles in spindle assembly, function, and orientation. Many acentrosomal cells exhibit prolonged spindle assembly, chromosome missegregation, DNA damage, misoriented divisions, and eventual apoptosis. We found that multiple mechanisms buffer the effects of centrosome loss, including alternative microtubule nucleation pathways and the spindle assembly checkpoint. Apoptosis of acentrosomal cells is mediated by JNK signaling, which also drives compensatory proliferation to maintain tissue integrity and viability. These data reveal the importance of centrosomes in fly epithelia and demonstrate the robust compensatory mechanisms at the cellular and organismal level.
Collapse
|
108
|
Gollin SM. Cytogenetic alterations and their molecular genetic correlates in head and neck squamous cell carcinoma: a next generation window to the biology of disease. Genes Chromosomes Cancer 2014; 53:972-90. [PMID: 25183546 DOI: 10.1002/gcc.22214] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 08/15/2014] [Indexed: 01/14/2023] Open
Abstract
Cytogenetic alterations underlie the development of head and neck squamous cell carcinoma (HNSCC), whether tobacco and alcohol use, betel nut chewing, snuff or human papillomavirus (HPV) causes the disease. Many of the molecular genetic aberrations in HNSCC result from these cytogenetic alterations. This review presents a brief introduction to the epidemiology of HNSCC, and discusses the role of HPV in the disease, cytogenetic alterations and their frequencies in HNSCC, their molecular genetic and The Cancer Genome Atlas (TCGA) correlates, prognostic implications, and possible therapeutic considerations. The most frequent cytogenetic alterations in HNSCC are gains of 5p14-15, 8q11-12, and 20q12-13, gains or amplifications of 3q26, 7p11, 8q24, and 11q13, and losses of 3p, 4q35, 5q12, 8p23, 9p21-24, 11q14-23, 13q12-14, 18q23, and 21q22. To understand their effects on tumor cell biology and response to therapy, the cytogenetic findings in HNSCC are increasingly being examined in the context of the biochemical pathways they disrupt. The goal is to minimize morbidity and mortality from HNSCC using cytogenetic abnormalities to identify valuable diagnostic biomarkers for HNSCC, prognostic biomarkers of tumor behavior, recurrence risk, and outcome, and predictive biomarkers of therapeutic response to identify the most efficacious treatment for each individual patient's tumor, all based on a detailed understanding of the next generation biology of HNSCC.
Collapse
Affiliation(s)
- Susanne M Gollin
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA; Departments of Otolaryngology and Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA; University of Pittsburgh Cancer Institute, Pittsburgh, PA
| |
Collapse
|
109
|
Shamanna RA, Singh DK, Lu H, Mirey G, Keijzers G, Salles B, Croteau DL, Bohr VA. RECQ helicase RECQL4 participates in non-homologous end joining and interacts with the Ku complex. Carcinogenesis 2014; 35:2415-24. [PMID: 24942867 DOI: 10.1093/carcin/bgu137] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
RECQL4, a member of the RecQ helicase family, is a multifunctional participant in DNA metabolism. RECQL4 protein participates in several functions both in the nucleus and in the cytoplasm of the cell, and mutations in human RECQL4 are associated with three genetic disorders: Rothmund-Thomson, RAPADILINO and Baller-Gerold syndromes. We previously reported that RECQL4 is recruited to laser-induced DNA double-strand breaks (DSB). Here, we have characterized the functional roles of RECQL4 in the non-homologous end joining (NHEJ) pathway of DSB repair. In an in vitro NHEJ assay that depends on the activity of DNA-dependent protein kinase (DNA-PK), extracts from RECQL4 knockdown cells display reduced end-joining activity on DNA substrates with cohesive and non-cohesive ends. Depletion of RECQL4 also reduced the end joining activity on a GFP reporter plasmid in vivo. Knockdown of RECQL4 increased the sensitivity of cells to γ-irradiation and resulted in accumulation of 53BP1 foci after irradiation, indicating defects in the processing of DSB. We find that RECQL4 interacts with the Ku70/Ku80 heterodimer, part of the DNA-PK complex, via its N-terminal domain. Further, RECQL4 stimulates higher order DNA binding of Ku70/Ku80 to a blunt end DNA substrate. Taken together, these results implicate that RECQL4 participates in the NHEJ pathway of DSB repair via a functional interaction with the Ku70/Ku80 complex. This is the first study to provide both in vitro and in vivo evidence for a role of a RecQ helicase in NHEJ.
Collapse
Affiliation(s)
- Raghavendra A Shamanna
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, 251 Bayview Boulevard, Baltimore, MD 21224, USA, INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Dharmendra Kumar Singh
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, 251 Bayview Boulevard, Baltimore, MD 21224, USA, INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Huiming Lu
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, 251 Bayview Boulevard, Baltimore, MD 21224, USA, INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Gladys Mirey
- INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and
| | - Guido Keijzers
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bernard Salles
- INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, 251 Bayview Boulevard, Baltimore, MD 21224, USA, INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, 251 Bayview Boulevard, Baltimore, MD 21224, USA, INRA, Université de Toulouse, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France and Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
110
|
The SUMO-targeted ubiquitin ligase RNF4 localizes to etoposide-exposed mitotic chromosomes: implication for a novel DNA damage response during mitosis. Biochem Biophys Res Commun 2014; 447:83-8. [PMID: 24695317 DOI: 10.1016/j.bbrc.2014.03.106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 03/21/2014] [Indexed: 11/24/2022]
Abstract
RNF4, a SUMO-targeted ubiquitin ligase (STUbL), localizes to the nucleus and functions in the DNA damage response during interphase of the cell cycle. RNF4 also exists in cells undergoing mitosis, where its regulation and function remain poorly understood. Here we showed that administration of etoposide, an anticancer DNA topoisomerase II poison, to mitotic human cervical cancer HeLa cells induced SUMO-2/3-dependent localization of RNF4 to chromosomes. The FK2 antibody signals, indicative of poly/multi-ubiquitin assembly, were detected on etoposide-exposed mitotic chromosomes, whereas the signals were negligible in cells depleted for RNF4 by RNA interference. This suggests that RNF4 functions as a STUbL in the etoposide-induced damage response during mitosis. Indeed, RNF4-depletion sensitized mitotic HeLa cells to etoposide and increased cells with micronuclei. These results indicate the importance of the RNF4-mediated STUbL pathway during mitosis for the maintenance of chromosome integrity and further implicate RNF4 as a target for topo II poison-based therapy for cancer patients.
Collapse
|
111
|
Voua Otomo P, Reinecke SA, Reinecke AJ. Using the comet assay to assess the combined and separate genotoxic effects of Cd and Zn in Eisenia andrei (Oligochaeta) at different temperatures. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2014; 92:285-288. [PMID: 24233261 DOI: 10.1007/s00128-013-1157-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/07/2013] [Indexed: 06/02/2023]
Abstract
Using the comet assay, the genotoxicity of Cd, Zn and Cd/Zn mixtures in Eisenia andrei was assessed after 4 weeks of exposure at 15, 20 and 25 °C. Relative to the controls, significant increases in TDNA% were observed in exposures to Cd alone at 500 and 1,000 mg/kg soil at both 20 and 25 °C, while a general decrease occurred at 15 °C. For Zn alone, a decreasing trend in TDNA% occurred at all three temperatures with increasing Zn concentration. For the Cd/Zn mixtures at 15 °C, genotoxicity was reduced at all mixture concentrations relative to the control. At 20 °C, the genotoxic response was similar to the control at all exposures. At 25 °C, the response was elevated at the 50 + 50 and 250 + 250 mg/kg mixture concentrations. In the remaining treatments at 25 °C, TDNA% was similar to the values in the respective control. The lack of consistently significant mixture genotoxicity may indicate antagonistic interactions between Cd and Zn in the mixtures. However, this was not conclusively determined because temperature alone had an inconsistent effect upon TDNA% readings in the control exposures.
Collapse
|
112
|
Genotoxic anti-cancer agents and their relationship to DNA damage, mitosis, and checkpoint adaptation in proliferating cancer cells. Int J Mol Sci 2014; 15:3403-31. [PMID: 24573252 PMCID: PMC3975345 DOI: 10.3390/ijms15033403] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/22/2014] [Accepted: 02/14/2014] [Indexed: 12/19/2022] Open
Abstract
When a human cell detects damaged DNA, it initiates the DNA damage response (DDR) that permits it to repair the damage and avoid transmitting it to daughter cells. Despite this response, changes to the genome occur and some cells, such as proliferating cancer cells, are prone to genome instability. The cellular processes that lead to genomic changes after a genotoxic event are not well understood. Our research focuses on the relationship between genotoxic cancer drugs and checkpoint adaptation, which is the process of mitosis with damaged DNA. We examine the types of DNA damage induced by widely used cancer drugs and describe their effects upon proliferating cancer cells. There is evidence that cell death caused by genotoxic cancer drugs in some cases includes exiting a DNA damage cell cycle arrest and entry into mitosis. Furthermore, some cells are able to survive this process at a time when the genome is most susceptible to change or rearrangement. Checkpoint adaptation is poorly characterised in human cells; we predict that increasing our understanding of this pathway may help to understand genomic instability in cancer cells and provide insight into methods to improve the efficacy of current cancer therapies.
Collapse
|
113
|
Gentric G, Desdouets C. Polyploidization in liver tissue. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:322-31. [PMID: 24140012 DOI: 10.1016/j.ajpath.2013.06.035] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/17/2013] [Accepted: 06/20/2013] [Indexed: 12/14/2022]
Abstract
Polyploidy (alias whole genome amplification) refers to organisms containing more than two basic sets of chromosomes. Polyploidy was first observed in plants more than a century ago, and it is known that such processes occur in many eukaryotes under a variety of circumstances. In mammals, the development of polyploid cells can contribute to tissue differentiation and, therefore, possibly a gain of function; alternately, it can be associated with development of disease, such as cancer. Polyploidy can occur because of cell fusion or abnormal cell division (endoreplication, mitotic slippage, or cytokinesis failure). Polyploidy is a common characteristic of the mammalian liver. Polyploidization occurs mainly during liver development, but also in adults with increasing age or because of cellular stress (eg, surgical resection, toxic exposure, or viral infections). This review will explore the mechanisms that lead to the development of polyploid cells, our current state of understanding of how polyploidization is regulated during liver growth, and its consequence on liver function.
Collapse
Affiliation(s)
- Géraldine Gentric
- French Institute of Health and Medical Research (INSERM), U1016, Cochin Institute, Department of Development, Reproduction and Cancer, Paris, France; French National Centre for Scientific Research (CNRS), UMR 8104, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Chantal Desdouets
- French Institute of Health and Medical Research (INSERM), U1016, Cochin Institute, Department of Development, Reproduction and Cancer, Paris, France; French National Centre for Scientific Research (CNRS), UMR 8104, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
114
|
Replication stress and mitotic dysfunction in cells expressing simian virus 40 large T antigen. J Virol 2013; 87:13179-92. [PMID: 24067972 DOI: 10.1128/jvi.02224-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously demonstrated that simian virus 40 (SV40) large T antigen (LT) binds to the Bub1 kinase, a key regulator of the spindle checkpoint and chromosome segregation. Bub1 mutations or altered expression patterns are linked to chromosome missegregation and are considered to be a driving force in some human cancers. Here we report that LT, dependent on Bub1 binding, causes micronuclei, lagging chromatin, and anaphase bridges, which are hallmarks of chromosomal instability (CIN) and Bub1 insufficiency. Using time-lapse microscopy, we demonstrate that LT imposes a Bub1 binding-dependent delay in the metaphase-to-anaphase transition. Kinetochore fibers reveal that LT, via Bub1 binding, causes aberrant kinetochore (KT)-microtubule (MT) attachments and a shortened interkinetochore distance, consistent with a lack of tension. Previously, we showed that LT also induces the DNA damage response (DDR) via Bub1 binding. Using inducible LT cell lines, we show that an activated DDR was observed before the appearance of anaphase bridges and micronuclei. Furthermore, LT induction in serum-starved cells demonstrated γ-H2AX accumulation in cells that had not yet entered mitosis. Thus, DDR activation can occur independently of chromosome segregation defects. Replication stress pathways may be responsible, because signatures of replication stress were observed, which were attenuated by exogenous supplementation with nucleosides. Our observations allow us to propose a model that explains and integrates the diverse manifestations of genomic instability induced by LT.
Collapse
|
115
|
Kofman AV, Kim J, Park SY, Dupart E, Letson C, Bao Y, Ding K, Chen Q, Schiff D, Larner J, Abounader R. microRNA-34a promotes DNA damage and mitotic catastrophe. Cell Cycle 2013; 12:3500-11. [PMID: 24091633 DOI: 10.4161/cc.26459] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Efficient and error-free DNA repair is critical for safeguarding genome integrity, yet it is also linked to radio- and chemoresistance of malignant tumors. miR-34a, a potent tumor suppressor, influences a large set of p53-regulated genes and contributes to p53-mediated apoptosis. However, the effects of miR-34a on the processes of DNA damage and repair are not entirely understood. We explored tet-inducible miR-34a-expressing human p53 wild-type and R273H p53 mutant GBM cell lines, and found that miR-34a influences the broad spectrum of 53BP1-mediated DNA damage response. It escalates both post-irradiation and endogenous DNA damage, abrogates radiation-induced G 2/M arrest and drastically increases the number of irradiated cells undergoing mitotic catastrophe. Furthermore, miR-34a downregulates 53BP1 and inhibits its recruitment to the sites of DNA double-strand breaks. We conclude that whereas miR-34a counteracts DNA repair, it also contributes to the p53-independent elimination of distressed cells, thus preventing the rise of genomic instability in tumor cell populations. These properties of miR-34a can potentially be exploited for DNA damage-effecting therapies of malignancies.
Collapse
Affiliation(s)
- Alexander V Kofman
- Department of Microbiology, Immunology and Cancer Biology; University of Virginia; Charlottesville, VA USA; Aging-Cancer Interface Group; LDS Medical Center; St. Petersburg, Russia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Orr B, Compton DA. A double-edged sword: how oncogenes and tumor suppressor genes can contribute to chromosomal instability. Front Oncol 2013; 3:164. [PMID: 23825799 PMCID: PMC3695391 DOI: 10.3389/fonc.2013.00164] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/06/2013] [Indexed: 12/21/2022] Open
Abstract
Most solid tumors are characterized by abnormal chromosome numbers (aneuploidy) and karyotypic profiling has shown that the majority of these tumors are heterogeneous and chromosomally unstable. Chromosomal instability (CIN) is defined as persistent mis-segregation of whole chromosomes and is caused by defects during mitosis. Large-scale genome sequencing has failed to reveal frequent mutations of genes encoding proteins involved in mitosis. On the contrary, sequencing has revealed that most mutated genes in cancer fall into a limited number of core oncogenic signaling pathways that regulate the cell cycle, cell growth, and apoptosis. This led to the notion that the induction of oncogenic signaling is a separate event from the loss of mitotic fidelity, but a growing body of evidence suggests that oncogenic signaling can deregulate cell cycle progression, growth, and differentiation as well as cause CIN. These new results indicate that the induction of CIN can no longer be considered separately from the cancer-associated driver mutations. Here we review the primary causes of CIN in mitosis and discuss how the oncogenic activation of key signal transduction pathways contributes to the induction of CIN.
Collapse
Affiliation(s)
- Bernardo Orr
- Department of Biochemistry, Geisel School of Medicine at Dartmouth , Hanover, NH , USA ; The Norris-Cotton Cancer Center, Geisel School of Medicine at Dartmouth , Hanover, NH , USA
| | | |
Collapse
|