1
|
Moon JI, Kim WJ, Kim KT, Kim HJ, Shin HR, Yoon H, Park SG, Park MS, Cho YD, Kim PJ, Ryoo HM. Foci-Xpress: Automated and Fast Nuclear Foci Counting Tool. Int J Mol Sci 2023; 24:14465. [PMID: 37833912 PMCID: PMC10572366 DOI: 10.3390/ijms241914465] [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: 08/10/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
In the nucleus, distinct, discrete spots or regions called "foci" have been identified, each harboring a specific molecular function. Accurate and efficient quantification of these foci is essential for understanding cellular dynamics and signaling pathways. In this study, we present an innovative automated image analysis method designed to precisely quantify subcellular foci within the cell nucleus. Manual foci counting methods can be tedious and time-consuming. To address these challenges, we developed an open-source software that automatically counts the number of foci from the indicated image files. We compared the foci counting efficiency, velocity, accuracy, and convenience of Foci-Xpress with those of other conventional methods in foci-induced models. We can adjust the brightness of foci to establish a threshold. The Foci-Xpress method was significantly faster than other conventional methods. Its accuracy was similar to that of conventional methods. The most significant strength of Foci-Xpress is automation, which eliminates the need for analyzing equipment while counting. This enhanced throughput facilitates comprehensive statistical analyses and supports robust conclusions from experiments. Furthermore, automation completely rules out biases caused by researchers, such as manual errors or daily variations. Thus, Foci-Xpress is a convincing, convenient, and easily accessible focus-counting tool for cell biologists.
Collapse
Affiliation(s)
- Jae-I Moon
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Woo-Jin Kim
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Ki-Tae Kim
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun-Jung Kim
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hye-Rim Shin
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Heein Yoon
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Gwa Park
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Min-Sang Park
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Dan Cho
- Department of Periodontology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea;
| | - Pil-Jong Kim
- Department of Biomedical Knowledge Engineering Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun-Mo Ryoo
- Department of Molecular Genetics and Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-Omics Center, Seoul National University, Seoul 08826, Republic of Korea; (J.-I.M.); (W.-J.K.); (K.-T.K.); (H.-J.K.); (H.-R.S.); (H.Y.); (S.G.P.); (M.-S.P.)
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
2
|
Haccard O, Ciardo D, Narrissamprakash H, Bronchain O, Kumagai A, Dunphy WG, Goldar A, Marheineke K. Rif1 restrains the rate of replication origin firing in Xenopus laevis. Commun Biol 2023; 6:788. [PMID: 37516798 PMCID: PMC10387115 DOI: 10.1038/s42003-023-05172-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023] Open
Abstract
Metazoan genomes are duplicated by the coordinated activation of clusters of replication origins at different times during S phase, but the underlying mechanisms of this temporal program remain unclear during early development. Rif1, a key replication timing factor, inhibits origin firing by recruiting protein phosphatase 1 (PP1) to chromatin counteracting S phase kinases. We have previously described that Rif1 depletion accelerates early Xenopus laevis embryonic cell cycles. Here, we find that in the absence of Rif1, patterns of replication foci change along with the acceleration of replication cluster activation. However, initiations increase only moderately inside active clusters. Our numerical simulations suggest that the absence of Rif1 compresses the temporal program towards more homogeneity and increases the availability of limiting initiation factors. We experimentally demonstrate that Rif1 depletion increases the chromatin-binding of the S phase kinase Cdc7/Drf1, the firing factors Treslin, MTBP, Cdc45, RecQL4, and the phosphorylation of both Treslin and MTBP. We show that Rif1 globally, but not locally, restrains the replication program in early embryos, possibly by inhibiting or excluding replication factors from chromatin.
Collapse
Affiliation(s)
- Olivier Haccard
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Diletta Ciardo
- Institut de Biologie de l'Ecole Normale Supérieure, Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Hemalatha Narrissamprakash
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Odile Bronchain
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris-Saclay, CERTO-Retina France, 91400, Saclay, France
| | - Akiko Kumagai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - William G Dunphy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Arach Goldar
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Kathrin Marheineke
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| |
Collapse
|
3
|
Lin K, Gueble SE, Sundaram RK, Huseman ED, Bindra RS, Herzon SB. Mechanism-based design of agents that selectively target drug-resistant glioma. Science 2022; 377:502-511. [PMID: 35901163 PMCID: PMC9502022 DOI: 10.1126/science.abn7570] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Approximately half of glioblastoma and more than two-thirds of grade II and III glioma tumors lack the DNA repair protein O6-methylguanine methyl transferase (MGMT). MGMT-deficient tumors respond initially to the DNA methylation agent temozolomide (TMZ) but frequently acquire resistance through loss of the mismatch repair (MMR) pathway. We report the development of agents that overcome this resistance mechanism by inducing MMR-independent cell killing selectively in MGMT-silenced tumors. These agents deposit a dynamic DNA lesion that can be reversed by MGMT but slowly evolves into an interstrand cross-link in MGMT-deficient settings, resulting in MMR-independent cell death with low toxicity in vitro and in vivo. This discovery may lead to new treatments for gliomas and may represent a new paradigm for designing chemotherapeutics that exploit specific DNA repair defects.
Collapse
Affiliation(s)
- Kingson Lin
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Susan E. Gueble
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ranjini K. Sundaram
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Eric D. Huseman
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Corresponding author. (S.B.H.); (R.S.B.)
| | - Seth B. Herzon
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, USA
- Corresponding author. (S.B.H.); (R.S.B.)
| |
Collapse
|
4
|
Malewicz NM, Rattray Z, Oeck S, Jung S, Escamilla-Rivera V, Chen Z, Tang X, Zhou J, LaMotte RH. Topical Capsaicin in Poly(lactic-co-glycolic)acid (PLGA) Nanoparticles Decreases Acute Itch and Heat Pain. Int J Mol Sci 2022; 23:5275. [PMID: 35563669 PMCID: PMC9101161 DOI: 10.3390/ijms23095275] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Capsaicin, the hot pepper agent, produces burning followed by desensitization. To treat localized itch or pain with minimal burning, low capsaicin concentrations can be repeatedly applied. We hypothesized that alternatively controlled release of capsaicin from poly(lactic-co-glycolic acid) (PLGA) nanoparticles desensitizes superficially terminating nociceptors, reducing burning. METHODS Capsaicin-loaded PLGA nanoparticles were prepared (single-emulsion solvent evaporation) and characterized (size, morphology, capsaicin loading, encapsulation efficiency, in vitro release profile). Capsaicin-PLGA nanoparticles were applied to murine skin and evaluated in healthy human participants (n = 21) for 4 days under blinded conditions, and itch and nociceptive sensations evoked by mechanical, heat stimuli and pruritogens cowhage, β-alanine, BAM8-22 and histamine were evaluated. RESULTS Nanoparticles (loading: 58 µg capsaicin/mg) released in vitro 23% capsaicin within the first hour and had complete release at 72 h. In mice, 24 h post-application Capsaicin-PLGA nanoparticles penetrated the dermis and led to decreased nociceptive behavioral responses to heat and mechanical stimulation (desensitization). Application in humans produced a weak to moderate burning, dissipating after 3 h. A loss of heat pain up to 2 weeks was observed. After capsaicin nanoparticles, itch and nociceptive sensations were reduced in response to pruritogens cowhage, β-alanine or BAM8-22, but were normal to histamine. CONCLUSIONS Capsaicin nanoparticles could be useful in reducing pain and itch associated with pruritic diseases that are histamine-independent.
Collapse
Affiliation(s)
- Nathalie M. Malewicz
- Department of Anesthesiology, Yale University School of Medicine, 330 Cedar St, New Haven, CT 06510, USA; (S.J.); (V.E.-R.)
- Clinics for Anesthesiology, Intensive Care and Pain Medicine, Medical Faculty of Ruhr-University Bochum, BG University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| | - Sebastian Oeck
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510, USA;
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Sebastian Jung
- Department of Anesthesiology, Yale University School of Medicine, 330 Cedar St, New Haven, CT 06510, USA; (S.J.); (V.E.-R.)
- ZEMOS Center for Solvation Science, Ruhr University Bochum, 44801 Bochum, Germany
| | - Vicente Escamilla-Rivera
- Department of Anesthesiology, Yale University School of Medicine, 330 Cedar St, New Haven, CT 06510, USA; (S.J.); (V.E.-R.)
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Zeming Chen
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA; (Z.C.); (X.T.); (J.Z.)
| | - Xiangjun Tang
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA; (Z.C.); (X.T.); (J.Z.)
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA; (Z.C.); (X.T.); (J.Z.)
| | - Robert H. LaMotte
- Department of Anesthesiology, Yale University School of Medicine, 330 Cedar St, New Haven, CT 06510, USA; (S.J.); (V.E.-R.)
| |
Collapse
|
5
|
A deep learning model (FociRad) for automated detection of γ-H2AX foci and radiation dose estimation. Sci Rep 2022; 12:5527. [PMID: 35365702 PMCID: PMC8975967 DOI: 10.1038/s41598-022-09180-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
DNA double-strand breaks (DSBs) are the most lethal form of damage to cells from irradiation. γ-H2AX (phosphorylated form of H2AX histone variant) has become one of the most reliable and sensitive biomarkers of DNA DSBs. However, the γ-H2AX foci assay still has limitations in the time consumed for manual scoring and possible variability between scorers. This study proposed a novel automated foci scoring method using a deep convolutional neural network based on a You-Only-Look-Once (YOLO) algorithm to quantify γ-H2AX foci in peripheral blood samples. FociRad, a two-stage deep learning approach, consisted of mononuclear cell (MNC) and γ-H2AX foci detections. Whole blood samples were irradiated with X-rays from a 6 MV linear accelerator at 1, 2, 4 or 6 Gy. Images were captured using confocal microscopy. Then, dose-response calibration curves were established and implemented with unseen dataset. The results of the FociRad model were comparable with manual scoring. MNC detection yielded 96.6% accuracy, 96.7% sensitivity and 96.5% specificity. γ-H2AX foci detection showed very good F1 scores (> 0.9). Implementation of calibration curve in the range of 0-4 Gy gave mean absolute difference of estimated doses less than 1 Gy compared to actual doses. In addition, the evaluation times of FociRad were very short (< 0.5 min per 100 images), while the time for manual scoring increased with the number of foci. In conclusion, FociRad was the first automated foci scoring method to use a YOLO algorithm with high detection performance and fast evaluation time, which opens the door for large-scale applications in radiation triage.
Collapse
|
6
|
Kaushik Tiwari M, Colon-Rios DA, Tumu HCR, Liu Y, Quijano E, Krysztofiak A, Chan C, Song E, Braddock DT, Suh HW, Saltzman WM, Rogers FA. Direct targeting of amplified gene loci for proapoptotic anticancer therapy. Nat Biotechnol 2022; 40:325-334. [PMID: 34711990 PMCID: PMC8930417 DOI: 10.1038/s41587-021-01057-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 08/05/2021] [Indexed: 12/16/2022]
Abstract
Gene amplification drives oncogenesis in a broad spectrum of cancers. A number of drugs have been developed to inhibit the protein products of amplified driver genes, but their clinical efficacy is often hampered by drug resistance. Here, we introduce a therapeutic strategy for targeting cancer-associated gene amplifications by activating the DNA damage response with triplex-forming oligonucleotides (TFOs), which drive the induction of apoptosis in tumors, whereas cells without amplifications process lower levels of DNA damage. Focusing on cancers driven by HER2 amplification, we find that TFOs targeting HER2 induce copy number-dependent DNA double-strand breaks (DSBs) and activate p53-independent apoptosis in HER2-positive cancer cells and human tumor xenografts via a mechanism that is independent of HER2 cellular function. This strategy has demonstrated in vivo efficacy comparable to that of current precision medicines and provided a feasible alternative to combat drug resistance in HER2-positive breast and ovarian cancer models. These findings offer a general strategy for targeting tumors with amplified genomic loci.
Collapse
Affiliation(s)
- Meetu Kaushik Tiwari
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Daniel A Colon-Rios
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Hemanta C Rao Tumu
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Elias Quijano
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
| | - Adam Krysztofiak
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Cynthia Chan
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Eric Song
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
| | | | - Hee-Won Suh
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
- Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Faye A Rogers
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Vicar T, Gumulec J, Kolar R, Kopecna O, Pagacova E, Falkova I, Falk M. DeepFoci: Deep learning-based algorithm for fast automatic analysis of DNA double-strand break ionizing radiation-induced foci. Comput Struct Biotechnol J 2022; 19:6465-6480. [PMID: 34976305 PMCID: PMC8668444 DOI: 10.1016/j.csbj.2021.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 11/26/2022] Open
Abstract
DNA double-strand breaks (DSBs), marked by ionizing radiation-induced (repair) foci (IRIFs), are the most serious DNA lesions and are dangerous to human health. IRIF quantification based on confocal microscopy represents the most sensitive and gold-standard method in radiation biodosimetry and allows research on DSB induction and repair at the molecular and single-cell levels. In this study, we introduce DeepFoci - a deep learning-based fully automatic method for IRIF counting and morphometric analysis. DeepFoci is designed to work with 3D multichannel data (trained for 53BP1 and γH2AX) and uses U-Net for nucleus segmentation and IRIF detection, together with maximally stable extremal region-based IRIF segmentation. The proposed method was trained and tested on challenging datasets consisting of mixtures of nonirradiated and irradiated cells of different types and IRIF characteristics - permanent cell lines (NHDFs, U-87) and primary cell cultures prepared from tumors and adjacent normal tissues of head and neck cancer patients. The cells were dosed with 0.5-8 Gy γ-rays and fixed at multiple (0-24 h) postirradiation times. Under all circumstances, DeepFoci quantified the number of IRIFs with the highest accuracy among current advanced algorithms. Moreover, while the detection error of DeepFoci remained comparable to the variability between two experienced experts, the software maintained its sensitivity and fidelity across dramatically different IRIF counts per nucleus. In addition, information was extracted on IRIF 3D morphometric features and repair protein colocalization within IRIFs. This approach allowed multiparameter IRIF categorization of single- or multichannel data, thereby refining the analysis of DSB repair processes and classification of patient tumors, with the potential to identify specific cell subclones. The developed software improves IRIF quantification for various practical applications (radiotherapy monitoring, biodosimetry, etc.) and opens the door to advanced DSB focus analysis and, in turn, a better understanding of (radiation-induced) DNA damage and repair.
Collapse
Key Words
- 53BP1, P53-binding protein 1
- Biodosimetry
- CNN, convolutional neural network
- Confocal Microscopy
- Convolutional Neural Network
- DNA Damage and Repair
- DSB, DNA double-strand break
- Deep Learning
- FOV, field of view
- GUI, graphical user interface
- IRIF, ionizing radiation-induced (repair) foci
- Image Analysis
- Ionizing Radiation-Induced Foci (IRIFs)
- MSER, maximally stable extremal region (algorithm)
- Morphometry
- NHDFs, normal human dermal fibroblasts
- RAD51, DNA repair protein RAD51 homolog 1
- U-87, U-87 glioblastoma cell line
- γH2AX, histone H2AX phosphorylated at serine 139
Collapse
Affiliation(s)
- Tomas Vicar
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, Brno, Czech Republic.,Czech Academy of Sciences, Institute of Biophysics, v.v.i, Department of Cell Biology and Radiobiology, Kralovopolska 135, Brno, Czech Republic.,Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - Jaromir Gumulec
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - Radim Kolar
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3058/10, Brno, Czech Republic
| | - Olga Kopecna
- Czech Academy of Sciences, Institute of Biophysics, v.v.i, Department of Cell Biology and Radiobiology, Kralovopolska 135, Brno, Czech Republic
| | - Eva Pagacova
- Czech Academy of Sciences, Institute of Biophysics, v.v.i, Department of Cell Biology and Radiobiology, Kralovopolska 135, Brno, Czech Republic
| | - Iva Falkova
- Czech Academy of Sciences, Institute of Biophysics, v.v.i, Department of Cell Biology and Radiobiology, Kralovopolska 135, Brno, Czech Republic
| | - Martin Falk
- Czech Academy of Sciences, Institute of Biophysics, v.v.i, Department of Cell Biology and Radiobiology, Kralovopolska 135, Brno, Czech Republic
| |
Collapse
|
8
|
Krysztofiak A, Szymonowicz K, Hlouschek J, Xiang K, Waterkamp C, Larafa S, Goetting I, Vega-Rubin-de-Celis S, Theiss C, Matschke V, Hoffmann D, Jendrossek V, Matschke J. Metabolism of cancer cells commonly responds to irradiation by a transient early mitochondrial shutdown. iScience 2021; 24:103366. [PMID: 34825138 PMCID: PMC8603201 DOI: 10.1016/j.isci.2021.103366] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 09/01/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022] Open
Abstract
Cancer bioenergetics fuel processes necessary to maintain viability and growth under stress conditions. We hypothesized that cancer metabolism supports the repair of radiation-induced DNA double-stranded breaks (DSBs). We combined the systematic collection of metabolic and radiobiological data from a panel of irradiated cancer cell lines with mathematical modeling and identified a common metabolic response with impact on the DSB repair kinetics, including a mitochondrial shutdown followed by compensatory glycolysis and resumption of mitochondrial function. Combining ionizing radiation (IR) with inhibitors of the compensatory glycolysis or mitochondrial respiratory chain slowed mitochondrial recovery and DNA repair kinetics, offering an opportunity for therapeutic intervention. Mathematical modeling allowed us to generate new hypotheses on general and individual mechanisms of the radiation response with relevance to DNA repair and on metabolic vulnerabilities induced by cancer radiotherapy. These discoveries will guide future mechanistic studies for the discovery of metabolic targets for overcoming intrinsic or therapy-induced radioresistance.
Collapse
Affiliation(s)
- Adam Krysztofiak
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Klaudia Szymonowicz
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Julian Hlouschek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Kexu Xiang
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Christoph Waterkamp
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, 45117 Essen, Germany
| | - Safa Larafa
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Isabell Goetting
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Silvia Vega-Rubin-de-Celis
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, 45117 Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
9
|
Köcher S, Volquardsen J, Perugachi Heinsohn A, Petersen C, Roggenbuck D, Rothkamm K, Mansour WY. Fully automated counting of DNA damage foci in tumor cell culture: A matter of cell separation. DNA Repair (Amst) 2021; 102:103100. [PMID: 33812230 DOI: 10.1016/j.dnarep.2021.103100] [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: 01/11/2021] [Revised: 02/18/2021] [Accepted: 03/14/2021] [Indexed: 11/17/2022]
Abstract
Analysis and quantification of residual, unrepaired DNA double-strand breaks by detecting damage-associated γH2AX or 53BP1 foci is a promising approach to evaluate radiosensitivity or radiosensitization in tumor cells. Manual foci quantification by eye is well-established but unsatisfactory due to inconsistent foci numbers between different observers, lack of information about foci size and intensity and the time-consuming scoring process. Therefore, automated foci counting is an important goal. Several software solutions for automated foci counting in separately acquired fluorescence microscopy images have been established. The AKLIDES NUK technology by Medipan combines automated microscopy and image processing/ counting, enabling affordable high throughput foci analysis as a routine application. Using this machine, automated foci counting is well established for lymphocytes but has not yet been reported for adherent tumor cells with their irregularly shaped nuclei and heterogeneous foci textures. Here we aimed to use the AKLIDES NUK system for adherent tumor cells growing in clusters. We identified cell separation as a critical step to ensure fast and reliable automated nuclei detection. We validated our protocol for the fully automated quantification of (i) the IR-dose dependent increase and (ii) the ATM as well as PARP inhibitor-induced radiosensitization. Collectively, with this protocol the AKLIDES NUK system facilitates cost effective, fast and high throughput quantitative fluorescence microscopic analysis of DNA damage induced foci such as γH2AX and 53BP1 in adherent tumor cells.
Collapse
Affiliation(s)
- S Köcher
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - J Volquardsen
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Perugachi Heinsohn
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - C Petersen
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - D Roggenbuck
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Senftenberg, Germany
| | - K Rothkamm
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - W Y Mansour
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Tumor Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt; Mildred-Scheel Cancer Career Center HATRICs4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Ventura JA, Donoghue JF, Nowell CJ, Cann LM, Day LRJ, Smyth LML, Forrester HB, Rogers PAW, Crosbie JC. The γH2AX DSB marker may not be a suitable biodosimeter to measure the biological MRT valley dose. Int J Radiat Biol 2021; 97:642-656. [PMID: 33617395 DOI: 10.1080/09553002.2021.1893854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/31/2020] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE γH2AX biodosimetry has been proposed as an alternative dosimetry method for microbeam radiation therapy (MRT) because conventional dosimeters, such as ionization chambers, lack the spatial resolution required to accurately measure the MRT valley dose. Here we investigated whether γH2AX biodosimetry should be used to measure the biological valley dose of MRT-irradiated mammalian cells. MATERIALS AND METHODS We irradiated human skin fibroblasts and mouse skin flaps with synchrotron MRT and broad beam (BB) radiation. BB doses of 1-5 Gy were used to generate a calibration curve in order to estimate the biological MRT valley dose using the γH2AX assay. RESULTS Our key finding was that MRT induced a non-linear dose response compared to BB, where doses 2-3 times greater showed the same level of DNA DSB damage in the valley in cell and tissue studies. This indicates that γH2AX may not be an appropriate biodosimeter to estimate the biological valley doses of MRT-irradiated samples. We also established foci yields of 5.9 ± 0 . 04 and 27.4 ± 2 . 5 foci/cell/Gy in mouse skin tissue and human fibroblasts respectively, induced by BB. Using Monte Carlo simulations, a linear dose response was seen in cell and tissue studies and produced predicted peak-to-valley dose ratios (PVDRs) of ∼30 and ∼107 for human fibroblasts and mouse skin tissue respectively. CONCLUSIONS Our report highlights novel MRT radiobiology, attempts to explain why γH2AX may not be an appropriate biodosimeter and suggests further studies aimed at revealing the biological and cellular communication mechanisms that drive the normal tissue sparing effect, which is characteristic of MRT.
Collapse
Affiliation(s)
- Jessica A Ventura
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Jacqueline F Donoghue
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Leonie M Cann
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Liam R J Day
- School of Science, RMIT University, Melbourne, Australia
| | - Lloyd M L Smyth
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Helen B Forrester
- School of Science, RMIT University, Melbourne, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Peter A W Rogers
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | | |
Collapse
|
11
|
Huna A, Griveau A, Vindrieux D, Jaber S, Flaman JM, Goehrig D, Azzi L, Médard JJ, Djebali S, Hernandez-Vargas H, Dante R, Payen L, Marvel J, Bertolino P, Aubert S, Dubus P, Bernard D. PLA2R1 promotes DNA damage and inhibits spontaneous tumor formation during aging. Cell Death Dis 2021; 12:190. [PMID: 33594040 PMCID: PMC7887270 DOI: 10.1038/s41419-021-03468-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
Although aging is a major risk factor for most types of cancers, it is barely studied in this context. The transmembrane protein PLA2R1 (phospholipase A2 receptor) promotes cellular senescence, which can inhibit oncogene-induced tumor initiation. Functions and mechanisms of action of PLA2R1 during aging are largely unknown. In this study, we observed that old Pla2r1 knockout mice were more prone to spontaneously develop a wide spectrum of tumors compared to control littermates. Consistently, these knockout mice displayed increased Parp1, a master regulator of DNA damage repair, and decreased DNA damage, correlating with large human dataset analysis. Forced PLA2R1 expression in normal human cells decreased PARP1 expression, induced DNA damage and subsequent senescence, while the constitutive expression of PARP1 rescued cells from these PLA2R1-induced effects. Mechanistically, PARP1 expression is repressed by a ROS (reactive oxygen species)-Rb-dependent mechanism upon PLA2R1 expression. In conclusion, our results suggest that PLA2R1 suppresses aging-induced tumors by repressing PARP1, via a ROS-Rb signaling axis, and inducing DNA damage and its tumor suppressive responses.
Collapse
Affiliation(s)
- Anda Huna
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Audrey Griveau
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - David Vindrieux
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Sara Jaber
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Jean-Michel Flaman
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Delphine Goehrig
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Lamia Azzi
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux Cedex, France
| | - Jean-Jacques Médard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Sophia Djebali
- Centre International de Recherche en Infectiologie, Inserm U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Hector Hernandez-Vargas
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Robert Dante
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Léa Payen
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Jacqueline Marvel
- Centre International de Recherche en Infectiologie, Inserm U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Sébastien Aubert
- Institut de Pathologie, Centre de Biologie Pathologie, CHRU de Lille, Faculté de Médecine, Université de Lille, Lille Cedex, France
| | - Pierre Dubus
- INSERM U1053 Bordeaux Research in Translational Oncology, University of Bordeaux, Bordeaux Cedex, France
- Plateau cellules tissus, CHU de Bordeaux, Pessac, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France.
| |
Collapse
|
12
|
Differential immunomodulatory effect of PARP inhibition in BRCA1 deficient and competent tumor cells. Biochem Pharmacol 2020; 184:114359. [PMID: 33285109 DOI: 10.1016/j.bcp.2020.114359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 01/23/2023]
Abstract
Poly-ADP-ribose polymerase (PARP) inhibitors are active against cells and tumors with defects in homology-directed repair as a result of synthetic lethality. PARP inhibitors (PARPi) have been suggested to act by either catalytic inhibition or by PARP localization in chromatin. In this study, we treat BRCA1 mutant cells derived from a patient with triple negative breast cancer and control cells for three weeks with veliparib, a PARPi, to determine if treatment with this drug induces increased levels of mutations and/or an inflammatory response. We show that long-term treatment with PARPi induces an inflammatory response in HCC1937 BRCA1 mutant cells. The levels of chromatin-bound PARP1 in the BRCA1 mutant cells correlate with significant upregulation of inflammatory genes and activation of the cyclic GMP-AMP synthase (cGAS)/signaling effector stimulator of interferon genes (STING pathway). In contrast, an increased mutational load is induced in BRCA1-complemented cells treated with a PARPi. Our results suggest that long-term PARP inhibitor treatment may prime both BRCA1 mutant and wild-type tumors for positive responses to immune checkpoint blockade, but by different underlying mechanisms.
Collapse
|
13
|
Nikitaki Z, Pariset E, Sudar D, Costes SV, Georgakilas AG. In Situ Detection of Complex DNA Damage Using Microscopy: A Rough Road Ahead. Cancers (Basel) 2020; 12:E3288. [PMID: 33172046 PMCID: PMC7694657 DOI: 10.3390/cancers12113288] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Complexity of DNA damage is considered currently one if not the primary instigator of biological responses and determinant of short and long-term effects in organisms and their offspring. In this review, we focus on the detection of complex (clustered) DNA damage (CDD) induced for example by ionizing radiation (IR) and in some cases by high oxidative stress. We perform a short historical perspective in the field, emphasizing the microscopy-based techniques and methodologies for the detection of CDD at the cellular level. We extend this analysis on the pertaining methodology of surrogate protein markers of CDD (foci) colocalization and provide a unique synthesis of imaging parameters, software, and different types of microscopy used. Last but not least, we critically discuss the main advances and necessary future direction for the better detection of CDD, with important outcomes in biological and clinical setups.
Collapse
Affiliation(s)
- Zacharenia Nikitaki
- Physics Department, School of Applied Mathematical and Physical Sciences, DNA Damage Laboratory, National Technical University of Athens (NTUA), 15780 Zografou, Athens, Greece
| | - Eloise Pariset
- Space Biosciences Division, Radiation Biophysics Laboratory, NASA Ames Research Center, Moffett Field, CA 94035, USA; (E.P.); (S.V.C.)
- Universities Space Research Association (USRA), Mountain View, CA 94043, USA
| | - Damir Sudar
- Life Sciences Department, Quantitative Imaging Systems LLC, Portland, OR 97209, USA;
| | - Sylvain V. Costes
- Space Biosciences Division, Radiation Biophysics Laboratory, NASA Ames Research Center, Moffett Field, CA 94035, USA; (E.P.); (S.V.C.)
| | - Alexandros G. Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, DNA Damage Laboratory, National Technical University of Athens (NTUA), 15780 Zografou, Athens, Greece
| |
Collapse
|
14
|
Chen X, Xun D, Zheng R, Zhao L, Lu Y, Huang J, Wang R, Wang Y. Deep-Learning-Assisted Assessment of DNA Damage Based on Foci Images and Its Application in High-Content Screening of Lead Compounds. Anal Chem 2020; 92:14267-14277. [DOI: 10.1021/acs.analchem.0c03741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Xuechun Chen
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dejin Xun
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ruzhang Zheng
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yuqing Lu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jun Huang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Rui Wang
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
15
|
Caveolin-1 regulates the ASMase/ceramide-mediated radiation response of endothelial cells in the context of tumor-stroma interactions. Cell Death Dis 2020; 11:228. [PMID: 32273493 PMCID: PMC7145831 DOI: 10.1038/s41419-020-2418-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/21/2022]
Abstract
The integral membrane protein caveolin-1 (CAV1) plays a central role in radioresistance-mediating tumor–stroma interactions of advanced prostate cancer (PCa). Among the tumor–stroma, endothelial cells (EC) evolved as critical determinants of the radiation response. CAV1 deficiency in angiogenic EC was already shown to account for increased apoptosis rates of irradiated EC. This study explores the potential impact of differential CAV1 levels in EC on the acid sphingomyelinase (ASMase)/ceramide pathway as a key player in the regulation of EC apoptosis upon irradiation and cancer cell radioresistance. Enhanced apoptosis sensitivity of CAV1-deficient EC was associated with increased ASMase activity, ceramide generation, formation of large lipid platforms, and finally an altered p38 mitogen-activated protein kinase (MAPK)/heat-shock protein 27 (HSP27)/AKT (protein kinase B, PKB) signaling. CAV1-deficient EC increased the growth delay of LNCaP and PC3 PCa cells upon radiation treatment in direct 3D spheroid co-cultures. Exogenous C6 and C16 ceramide treatment in parallel increased the growth delay of PCa spheroids and induced PCa cell apoptosis. Analysis of the respective ceramide species in PCa cells with increased CAV1 levels like those typically found in radio-resistant advanced prostate tumors further revealed an upregulation of unsaturated C24:1 ceramide that might scavenge the effects of EC-derived apoptosis-inducing C16 ceramide. Higher ASMase as well as ceramide levels could be confirmed by immunohistochemistry in human advanced prostate cancer specimen bearing characteristic CAV1 tumor–stroma alterations. Conclusively, CAV1 critically regulates the generation of ceramide-dependent (re-)organization of the plasma membrane that in turn affects the radiation response of EC and adjacent PCa cells. Understanding the CAV1-dependent crosstalk between tumor cells and the host-derived tumor microvasculature and its impact on radiosensitivity may allow to define a rational strategy for overcoming tumor radiation resistance improving clinical outcomes by targeting CAV1.
Collapse
|
16
|
Szymonowicz K, Krysztofiak A, van der Linden J, Kern A, Deycmar S, Oeck S, Squire A, Koska B, Hlouschek J, Vüllings M, Neander C, Siveke JT, Matschke J, Pruschy M, Timmermann B, Jendrossek V. Proton Irradiation Increases the Necessity for Homologous Recombination Repair Along with the Indispensability of Non-Homologous End Joining. Cells 2020; 9:E889. [PMID: 32260562 PMCID: PMC7226794 DOI: 10.3390/cells9040889] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Technical improvements in clinical radiotherapy for maximizing cytotoxicity to the tumor while limiting negative impact on co-irradiated healthy tissues include the increasing use of particle therapy (e.g., proton therapy) worldwide. Yet potential differences in the biology of DNA damage induction and repair between irradiation with X-ray photons and protons remain elusive. We compared the differences in DNA double strand break (DSB) repair and survival of cells compromised in non-homologous end joining (NHEJ), homologous recombination repair (HRR) or both, after irradiation with an equal dose of X-ray photons, entrance plateau (EP) protons, and mid spread-out Bragg peak (SOBP) protons. We used super-resolution microscopy to investigate potential differences in spatial distribution of DNA damage foci upon irradiation. While DNA damage foci were equally distributed throughout the nucleus after X-ray photon irradiation, we observed more clustered DNA damage foci upon proton irradiation. Furthermore, deficiency in essential NHEJ proteins delayed DNA repair kinetics and sensitized cells to both, X-ray photon and proton irradiation, whereas deficiency in HRR proteins sensitized cells only to proton irradiation. We assume that NHEJ is indispensable for processing DNA DSB independent of the irradiation source, whereas the importance of HRR rises with increasing energy of applied irradiation.
Collapse
Affiliation(s)
- Klaudia Szymonowicz
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
| | - Adam Krysztofiak
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
| | - Jansje van der Linden
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
| | - Ajvar Kern
- West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany; (A.K.); (B.K.); (M.V.); (B.T.)
| | - Simon Deycmar
- Department of Radiation Oncology, Laboratory for Applied Radiobiology, University Hospital Zurich, Zurich, Switzerland; (S.D.); (M.P.)
| | - Sebastian Oeck
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Anthony Squire
- Institute of Experimental Immunology and Imaging, Imaging Center Essen, University Hospital Essen, 45122 Essen, Germany;
| | - Benjamin Koska
- West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany; (A.K.); (B.K.); (M.V.); (B.T.)
| | - Julian Hlouschek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
| | - Melanie Vüllings
- West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany; (A.K.); (B.K.); (M.V.); (B.T.)
| | - Christian Neander
- Institute of Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany; (C.N.); (J.T.S.)
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Jens T. Siveke
- Institute of Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany; (C.N.); (J.T.S.)
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
| | - Martin Pruschy
- Department of Radiation Oncology, Laboratory for Applied Radiobiology, University Hospital Zurich, Zurich, Switzerland; (S.D.); (M.P.)
| | - Beate Timmermann
- West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany; (A.K.); (B.K.); (M.V.); (B.T.)
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
- Department of Particle Therapy, West German Proton Therapy Center Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (K.S.); (A.K.); (J.v.d.L.); (S.O.); (J.H.); (J.M.)
| |
Collapse
|
17
|
Memmel S, Sisario D, Zimmermann H, Sauer M, Sukhorukov VL, Djuzenova CS, Flentje M. FocAn: automated 3D analysis of DNA repair foci in image stacks acquired by confocal fluorescence microscopy. BMC Bioinformatics 2020; 21:27. [PMID: 31992200 PMCID: PMC6986076 DOI: 10.1186/s12859-020-3370-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 01/15/2020] [Indexed: 11/29/2022] Open
Abstract
Background Phosphorylated histone H2AX, also known as γH2AX, forms μm-sized nuclear foci at the sites of DNA double-strand breaks (DSBs) induced by ionizing radiation and other agents. Due to their specificity and sensitivity, γH2AX immunoassays have become the gold standard for studying DSB induction and repair. One of these assays relies on the immunofluorescent staining of γH2AX followed by microscopic imaging and foci counting. During the last years, semi- and fully automated image analysis, capable of fast detection and quantification of γH2AX foci in large datasets of fluorescence images, are gradually replacing the traditional method of manual foci counting. A major drawback of the non-commercial software for foci counting (available so far) is that they are restricted to 2D-image data. In practice, these algorithms are useful for counting the foci located close to the midsection plane of the nucleus, while the out-of-plane foci are neglected. Results To overcome the limitations of 2D foci counting, we present a freely available ImageJ-based plugin (FocAn) for automated 3D analysis of γH2AX foci in z-image stacks acquired by confocal fluorescence microscopy. The image-stack processing algorithm implemented in FocAn is capable of automatic 3D recognition of individual cell nuclei and γH2AX foci, as well as evaluation of the total foci number per cell nucleus. The FocAn algorithm consists of two parts: nucleus identification and foci detection, each employing specific sequences of auto local thresholding in combination with watershed segmentation techniques. We validated the FocAn algorithm using fluorescence-labeled γH2AX in two glioblastoma cell lines, irradiated with 2 Gy and given up to 24 h post-irradiation for repair. We found that the data obtained with FocAn agreed well with those obtained with an already available software (FoCo) and manual counting. Moreover, FocAn was capable of identifying overlapping foci in 3D space, which ensured accurate foci counting even at high DSB density of up to ~ 200 DSB/nucleus. Conclusions FocAn is freely available an open-source 3D foci analyzer. The user-friendly algorithm FocAn requires little supervision and can automatically count the amount of DNA-DSBs, i.e. fluorescence-labeled γH2AX foci, in 3D image stacks acquired by laser-scanning microscopes without additional nuclei staining.
Collapse
Affiliation(s)
- Simon Memmel
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany
| | - Dmitri Sisario
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum, Universität Würzburg, 97074, Würzburg, Germany
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany.,Molekulare und Zellulare Biotechnologie/Nanotechnologie, Universität des Saarlandes, Campus Saarbrücken, 66123, Saarbrücken, Germany.,Marine Sciences, Universidad Catolica del Norte, Casa Central, Angamos 0610, Antafogasta/Coquimbo, Chile
| | - Markus Sauer
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum, Universität Würzburg, 97074, Würzburg, Germany
| | - Vladimir L Sukhorukov
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum, Universität Würzburg, 97074, Würzburg, Germany
| | - Cholpon S Djuzenova
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany.
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany.
| |
Collapse
|
18
|
Wu Z, Oeck S, West AP, Mangalhara KC, Sainz AG, Newman LE, Zhang XO, Wu L, Yan Q, Bosenberg M, Liu Y, Sulkowski PL, Tripple V, Kaech SM, Glazer PM, Shadel GS. Mitochondrial DNA Stress Signalling Protects the Nuclear Genome. Nat Metab 2019; 1:1209-1218. [PMID: 32395698 PMCID: PMC7213273 DOI: 10.1038/s42255-019-0150-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 11/08/2019] [Indexed: 12/20/2022]
Abstract
The mammalian genome comprises nuclear DNA (nDNA) derived from both parents and mitochondrial DNA (mtDNA) that is maternally inherited and encodes essential proteins required for oxidative phosphorylation. Thousands of copies of the circular mtDNA are present in most cell types that are packaged by TFAM into higher-order structures called nucleoids1. Mitochondria are also platforms for antiviral signalling2 and, due to their bacterial origin, mtDNA and other mitochondrial components trigger innate immune responses and inflammatory pathology2,3. We showed previously that instability and cytoplasmic release of mtDNA activates the cGAS-STING-TBK1 pathway resulting in interferon stimulated gene (ISG) expression that promotes antiviral immunity4. Here, we find that persistent mtDNA stress is not associated with basally activated NF-κB signalling or interferon gene expression typical of an acute antiviral response. Instead, a specific subset of ISGs, that includes Parp9, remains activated by the unphosphorylated form of ISGF3 (U-ISGF3) that enhances nDNA damage and repair responses. In cultured primary fibroblasts and cancer cells, the chemotherapeutic drug doxorubicin causes mtDNA damage and release, which leads to cGAS-STING-dependent ISG activation. In addition, mtDNA stress in TFAM-deficient mouse melanoma cells produces tumours that are more resistant to doxorubicin in vivo. Finally, Tfam +/- mice exposed to ionizing radiation exhibit enhanced nDNA repair responses in spleen. Therefore, we propose that damage to and subsequent release of mtDNA elicits a protective signalling response that enhances nDNA repair in cells and tissues, suggesting mtDNA is a genotoxic stress sentinel.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sebastian Oeck
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Essen, Germany
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M College of Medicine, Bryan, TX, USA
| | | | - Alva G Sainz
- Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Laura E Newman
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Xiao-Ou Zhang
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lizhen Wu
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Marcus Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Department of Immunology, Yale School of Medicine, New Haven, CT, USA
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Parker L Sulkowski
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Susan M Kaech
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Peter M Glazer
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
19
|
Gutiérrez-Corbo C, Álvarez-Velilla R, Reguera RM, García-Estrada C, Cushman M, Balaña-Fouce R, Pérez-Pertejo Y. Topoisomerase IB poisons induce histone H2A phosphorylation as a response to DNA damage in Leishmania infantum. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 11:39-48. [PMID: 31563118 PMCID: PMC6796659 DOI: 10.1016/j.ijpddr.2019.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023]
Abstract
DNA topoisomerases are considered consolidated druggable targets against diseases produced by trypanosomatids. Several reports indicated that indenoisoquinolines, a family of non-camptothecinic based topoisomerase poisons, have a strong leishmanicidal effect both in vitro and in vivo in murine models of visceral leishmaniasis. The antileishmanial effect of the indenoisoquinolines implies several mechanisms that include the stabilization of the cleavage complex, histone H2A phosphorylation and DNA fragmentation. A series of 20 compounds with the indenoisoquinoline scaffold and several substituents at positions N6, C3, C8 and C9, were tested both in promastigotes and in intramacrophage splenic amastigotes obtained from an experimental murine infection. The antileishmanial effect of most of these compounds was within the micromolar or submicromolar range. In addition, the introduction of an N atom in the indenoisoquinoline ring (7-azaindenoisoquinolines) produced the highest selectivity index along with strong DNA topoisomerase IB inhibition, histone H2A phosphorylation and DNA-topoisomerase IB complex stabilization. This report shows for the first time the effect of a series of synthetic indenoisoquinolines on histone H2A phosphorylation, which represents a primary signal of double stranded DNA break in genus Leishmania. N-6 indenoisoquinoline derivatives show strong antileishmanial activity. Indenoisoquinolines arrest Leishmania cell-cycle in S phase. Inhibition of leishmanial TopIB by indenoisoquinolines induces DNA fragmentation. Leishmanial H2A histone is phosphorylated at the Thr128 in response to DNA damage.
Collapse
Affiliation(s)
- Camino Gutiérrez-Corbo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana S/n, 24071, León, Spain
| | - Raquel Álvarez-Velilla
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana S/n, 24071, León, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana S/n, 24071, León, Spain
| | - Carlos García-Estrada
- INBIOTEC (Instituto de Biotecnología de León), Avda. Real 1 - Parque Científico de León, 24006, León, Spain
| | - Mark Cushman
- Department of Medicinal Chemistry, and Molecular Pharmacology, College of Pharmacy, The Purdue Center for Cancer Research, Purdue University, Lafayette, IN, USA
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana S/n, 24071, León, Spain
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana S/n, 24071, León, Spain.
| |
Collapse
|
20
|
Differences in itch and pain behaviors accompanying the irritant and allergic contact dermatitis produced by a contact allergen in mice. Pain Rep 2019; 4:e781. [PMID: 31875186 PMCID: PMC6882579 DOI: 10.1097/pr9.0000000000000781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/26/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. Introduction: Irritant contact dermatitis (ICD) and allergic contact dermatitis (ACD) are inflammatory skin diseases accompanied by itch and pain. Irritant contact dermatitis is caused by chemical irritants eliciting an innate immune response, whereas ACD is induced by haptens additionally activating an adaptive immune response: After initial exposure (sensitization) to the hapten, a subsequent challenge can lead to a delayed-type hypersensitivity reaction. But, the sensory and inflammatory effects of sensitization (ICD) vs challenge of ACD are insufficiently studied. Therefore, we compared itch- and pain-like behaviors and inflammatory reactions evoked in mice during the sensitization (ICD) vs challenge phase (ACD) of application of the hapten, squaric acid dibutylester (SADBE). Objectives: Our aim was to compare itch- and pain-like behaviors and inflammatory reactions evoked in mice during the sensitization (ICD) vs challenge phase (ACD) of application of the hapten, squaric acid dibutylester (SADBE). Methods: Mice were sensitized on the abdomen with 1% SADBE (ACD) or vehicle treated (ICD, control). Spontaneous and stimulus-evoked itch- and pain-like behaviors were recorded in mice before and after 3 daily challenges of the cheek with 1% SADBE (ACD, ICD). Cutaneous inflammation was evaluated with clinical scoring, ultrasound imaging, skin thickness, histology, and analyses of selected biomarkers for contact dermatitis, IL-1β, TNF-α, CXCL10, and CXCR3. Results: Allergic contact dermatitis vs ICD mice exhibited more spontaneous site-directed scratching (itch) and wiping (pain). Allergic contact dermatitis—but not ICD—mice exhibited allodynia and hyperalgesia to mechanical and heat stimuli. Inflammatory mediators IL-1β and TNF-α were upregulated in both groups as well as the chemokine receptor, CXCR3. CXCL10, a CXCR3 ligand, was upregulated only for ACD. Inflammatory responses were more pronounced in ACD than ICD. Conclusion: These findings provide new information for differentiating the behavioral and inflammatory reactions to hapten-induced ICD and ACD.
Collapse
|
21
|
Rassamegevanon T, Löck S, Baumann M, Krause M, von Neubeck C. Comparable radiation response of ex vivo and in vivo irradiated tumor samples determined by residual γH2AX. Radiother Oncol 2019; 139:94-100. [PMID: 31445839 DOI: 10.1016/j.radonc.2019.06.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/16/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE a) To investigate if an ex vivo cultured and irradiated tumor biopsy reflects and predicts the radiation response of the corresponding in vivo irradiated tumor measured with the DNA double strand break marker γH2AX foci. MATERIALS AND METHODS Five human head and neck squamous cell carcinoma (hHNSCC) xenograft models were used. Fine needle biopsies were taken from anesthetized tumor-bearing NMRI nude mice prior to in vivo single dose irradiation (0, 2, 4, or 8 Gy) under ambient blood flow. Biopsies were ex vivo reoxygenated and irradiated with equivalent doses. Tumors and biopsies were fixed 24 h post irradiation, and γH2AX foci were assessed in oxygenated tumor regions. RESULTS Linear regression analysis showed comparable slopes of the residual γH2AX foci dose-response curves in four out of five hHNSCC models when in vivo and ex vivo cohorts were compared. The slopes from ex vivo biopsies and in vivo tumors could classify the respective tumor model as sensitive or resistant according to the intrinsic radiation sensitivity (TCD50). CONCLUSION The ability of ex vivo irradiated tumor biopsies to reflect and predict the intrinsic radiation response of in vivo tumors increases the translational potential of the ex vivo γH2AX foci assay as a diagnostic tool for clinical practice.
Collapse
Affiliation(s)
- Treewut Rassamegevanon
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany.
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Cläre von Neubeck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
22
|
RB1 Deletion in Retinoblastoma Protein Pathway-Disrupted Cells Results in DNA Damage and Cancer Progression. Mol Cell Biol 2019; 39:MCB.00105-19. [PMID: 31138663 DOI: 10.1128/mcb.00105-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
Proliferative control in cancer cells is frequently disrupted by mutations in the retinoblastoma protein (RB) pathway. Intriguingly, RB1 mutations can arise late in tumorigenesis in cancer cells whose RB pathway is already compromised by another mutation. In this study, we present evidence for increased DNA damage and instability in cancer cells with RB pathway defects when RB1 mutations are induced. We generated isogenic RB1 mutant genotypes with CRISPR/Cas9 in a number of cell lines. Cells with even one mutant copy of RB1 have increased basal levels of DNA damage and increased mitotic errors. Elevated levels of reactive oxygen species as well as impaired homologous recombination repair underlie this DNA damage. When xenografted into immunocompromised mice, RB1 mutant cells exhibit an elevated propensity to seed new tumors in recipient lungs. This study offers evidence that late-arising RB1 mutations can facilitate genome instability and cancer progression that are beyond the preexisting proliferative control deficit.
Collapse
|
23
|
Barbieri S, Baiocco G, Babini G, Morini J, Friedland W, Buonanno M, Grilj V, Brenner DJ, Ottolenghi A. MODELLING γ-H2AX FOCI INDUCTION TO MIMIC LIMITATIONS IN THE SCORING TECHNIQUE. RADIATION PROTECTION DOSIMETRY 2019; 183:121-125. [PMID: 30520984 PMCID: PMC6525333 DOI: 10.1093/rpd/ncy217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
An approach based on track-structure calculations has been developed to take account of artefacts occurring during γ-H2AX foci detection in 2D images of samples analyzed through immunocytochemistry. The need of this works stems from the observed saturation in foci yields measured after X-ray doses higher than few grays, hindering an unambiguous quantification of DNA damage and of radiation effectiveness. The proposed modelling approach allows to simulate the observer's point of view for foci scoring, mimicking the selection of a slice Δz of the cell nucleus due to the microscope depth of field, and applying a clustering algorithm to group together damages within a resolution parameter r. Calculation results were benchmarked with experimental measurements at an early time-point for mouse breast cancer cells, irradiated with X-ray doses in the range 0-5 Gy. The model is able to reproduce the saturation in experimental data.
Collapse
Affiliation(s)
| | | | | | - Jacopo Morini
- Physics Department, University of Pavia, Pavia, Italy
| | - Werner Friedland
- Institute of Radiation Protection, Department of Radiation Sciences, Helmholtz Zentrum München—German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Manuela Buonanno
- Center for Radiological Research, Columbia University Medical Center, New York, New York, USA
| | - Veljko Grilj
- Center for Radiological Research, Columbia University Medical Center, New York, New York, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Medical Center, New York, New York, USA
| | | |
Collapse
|
24
|
Guard SE, Poss ZC, Ebmeier CC, Pagratis M, Simpson H, Taatjes DJ, Old WM. The nuclear interactome of DYRK1A reveals a functional role in DNA damage repair. Sci Rep 2019; 9:6539. [PMID: 31024071 PMCID: PMC6483993 DOI: 10.1038/s41598-019-42990-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/12/2019] [Indexed: 12/21/2022] Open
Abstract
The chromosome 21 encoded protein kinase DYRK1A is essential for normal human development. Mutations in DYRK1A underlie a spectrum of human developmental disorders, and increased dosage in trisomy 21 is implicated in Down syndrome related pathologies. DYRK1A regulates a diverse array of cellular processes through physical interactions with substrates and binding partners in various subcellular compartments. Despite recent large-scale protein-protein interaction profiling efforts, DYRK1A interactions specific to different subcellular compartments remain largely unknown, impeding progress toward understanding emerging roles for this kinase. Here, we used immunoaffinity purification and quantitative mass spectrometry to identify nuclear interaction partners of endogenous DYRK1A. This interactome was enriched in DNA damage repair factors, transcriptional elongation factors and E3 ubiquitin ligases. We validated an interaction with RNF169, a factor that promotes homology directed repair upon DNA damage, and found that DYRK1A expression and kinase activity are required for maintenance of 53BP1 expression and subsequent recruitment to DNA damage loci. Further, DYRK1A knock out conferred resistance to ionizing radiation in colony formation assays, suggesting that DYRK1A expression decreases cell survival efficiency in response to DNA damage and points to a tumor suppressive role for this kinase.
Collapse
Affiliation(s)
- Steven E Guard
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Zachary C Poss
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Christopher C Ebmeier
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Maria Pagratis
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Helen Simpson
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Dylan J Taatjes
- Department of Biochemistry, University of Colorado, Boulder, CO, USA
| | - William M Old
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
25
|
Oeck S, Malewicz NM, Krysztofiak A, Turchick A, Jendrossek V, Glazer PM. High-throughput Evaluation of Protein Migration and Localization after Laser Micro-Irradiation. Sci Rep 2019; 9:3148. [PMID: 30816253 PMCID: PMC6395640 DOI: 10.1038/s41598-019-39760-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/01/2019] [Indexed: 12/26/2022] Open
Abstract
DNA- and histone-related research frequently comprises the quantitative analysis of protein modifications, such as histone phosphorylation. Analysis of accumulation and disappearance of protein foci are used to monitor DNA damage and repair kinetics. If the protein of interest doesn't accumulate in foci, laser micro-irradiation of single nuclei provides an alternative method to monitor DNA repair proteins and histone dynamics at the DNA damage site. We have developed an automated evaluation tool for standardized, high-throughput analysis of micro-irradiated cells featuring single cell background subtraction and detection across multiple fluorescence channels, allowing for robust statistics.
Collapse
Affiliation(s)
- Sebastian Oeck
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, 45122, Germany.
| | - Nathalie M Malewicz
- Department of Anaesthesiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Adam Krysztofiak
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, 45122, Germany
| | - Audrey Turchick
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, 45122, Germany
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
26
|
Wiesemann A, Ketteler J, Slama A, Wirsdörfer F, Hager T, Röck K, Engel DR, Fischer JW, Aigner C, Jendrossek V, Klein D. Inhibition of Radiation-Induced Ccl2 Signaling Protects Lungs from Vascular Dysfunction and Endothelial Cell Loss. Antioxid Redox Signal 2019; 30:213-231. [PMID: 29463096 DOI: 10.1089/ars.2017.7458] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aims: Radiation-induced normal tissue toxicity often precludes the application of curative radiation doses. Here we investigated the therapeutic potential of chemokine C-C motif ligand 2 (Ccl2) signaling inhibition to protect normal lung tissue from radiotherapy (RT)-induced injury. Results: RT-induced vascular dysfunction and associated adverse effects can be efficiently antagonized by inhibition of Ccl2 signaling using either the selective Ccl2 inhibitor bindarit (BIN) or mice deficient for the main Ccl2 receptor CCR2 (KO). BIN-treatment efficiently counteracted the RT-induced expression of Ccl2, normalized endothelial cell (EC) morphology and vascular function, and limited lung inflammation and metastasis early after irradiation (acute effects). A similar protection of the vascular compartment was detected by loss of Ccl2 signaling in lungs of CCR2-KO mice. Long-term Ccl2 signaling inhibition also significantly limited EC loss and accompanied fibrosis progression as adverse late effect. With respect to the human situation, we further confirmed that Ccl2 secreted by RT-induced senescent epithelial cells resulted in the activation of normally quiescent but DNA-damaged EC finally leading to EC loss in ex vivo cultured human normal lung tissue. Innovation: Abrogation of certain aspects of the secretome of irradiated resident lung cells, in particular signaling inhibition of the senescence-associated secretory phenotype-factor Ccl2 secreted predominantly by RT-induced senescent epithelial cells, resulted in protection of the endothelial compartment. Conclusions: Radioprotection of the normal tissue via Ccl2 signaling inhibition without simultaneous protection or preferable radiosensitization of tumor tissue might improve local tumor control and survival, because higher doses of radiation could be used.
Collapse
Affiliation(s)
- Alina Wiesemann
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Julia Ketteler
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Alexis Slama
- 2 Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik-University Clinic Essen , Essen, Germany
| | - Florian Wirsdörfer
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Thomas Hager
- 3 Institute of Pathology, University Clinic Essen, University of Duisburg-Essen , Essen, Germany
| | - Katharina Röck
- 4 Institute for Pharmacology, University Hospital, Heinrich-Heine-University , Düsseldorf, Germany
| | - Daniel R Engel
- 5 Department Immunodynamics, Institute of Experimental Immunology and Imaging, University Duisburg-Essen, University Hospital Essen , Essen, Germany
| | - Jens W Fischer
- 4 Institute for Pharmacology, University Hospital, Heinrich-Heine-University , Düsseldorf, Germany
| | - Clemens Aigner
- 2 Department of Thoracic Surgery and Surgical Endoscopy, Ruhrlandklinik-University Clinic Essen , Essen, Germany
| | - Verena Jendrossek
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Diana Klein
- 1 Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital , Essen, Germany
| |
Collapse
|
27
|
Ma X, Warnier M, Raynard C, Ferrand M, Kirsh O, Defossez PA, Martin N, Bernard D. The nuclear receptor RXRA controls cellular senescence by regulating calcium signaling. Aging Cell 2018; 17:e12831. [PMID: 30216632 PMCID: PMC6260923 DOI: 10.1111/acel.12831] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 07/10/2018] [Accepted: 07/21/2018] [Indexed: 12/18/2022] Open
Abstract
Calcium signaling is emerging as a key pathway controlling cellular senescence, a stable cell proliferation arrest playing a fundamental role in pathophysiological conditions, such as embryonic development, wound healing, cancer, and aging. However, how calcium signaling is regulated is still only partially understood. The inositol 1, 4, 5‐trisphosphate receptor type 2 (ITPR2), an endoplasmic reticulum calcium release channel, was recently shown to critically contribute to the implementation of senescence, but how ITPR2 expression is controlled is unclear. To gain insights into the regulation of ITPR2 expression, we performed an siRNA screen targeting 160 transcription factors and epigenetic regulators. Interestingly, we discovered that the retinoid X receptor alpha (RXRA), which belongs to the nuclear receptor family, represses ITPR2 expression and regulates calcium signaling though ITPR2 and the mitochondrial calcium uniporter (MCU). Knockdown of RXRA induces the production of reactive oxygen species (ROS) and DNA damage via the ITPR2‐MCU calcium signaling axis and consequently triggers cellular senescence by activating p53, whereas RXRA overexpression decreases DNA damage accumulation and then delays replicative senescence. Altogether, our work sheds light on a novel mechanism controlling calcium signaling and cellular senescence and provides new insights into the role of nuclear receptors.
Collapse
Affiliation(s)
- Xingjie Ma
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Marine Warnier
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Clotilde Raynard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Mylène Ferrand
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - Olivier Kirsh
- Epigenetics and Cell Fate, Sorbonne Paris Cité, CNRS UMR 7216; Université Paris Diderot; Paris France
| | - Pierre-Antoine Defossez
- Epigenetics and Cell Fate, Sorbonne Paris Cité, CNRS UMR 7216; Université Paris Diderot; Paris France
| | - Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard; Université de Lyon; Lyon France
| |
Collapse
|
28
|
Oeck S, Szymonowicz K, Wiel G, Krysztofiak A, Lambert J, Koska B, Iliakis G, Timmermann B, Jendrossek V. Relating Linear Energy Transfer to the Formation and Resolution of DNA Repair Foci After Irradiation with Equal Doses of X-ray Photons, Plateau, or Bragg-Peak Protons. Int J Mol Sci 2018; 19:ijms19123779. [PMID: 30486506 PMCID: PMC6320817 DOI: 10.3390/ijms19123779] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 12/27/2022] Open
Abstract
Proton beam therapy is increasingly applied for the treatment of human cancer, as it promises to reduce normal tissue damage. However, little is known about the relationship between linear energy transfer (LET), the type of DNA damage, and cellular repair mechanisms, particularly for cells irradiated with protons. We irradiated cultured cells delivering equal doses of X-ray photons, Bragg-peak protons, or plateau protons and used this set-up to quantitate initial DNA damage (mainly DNA double strand breaks (DSBs)), and to analyze kinetics of repair by detecting γH2A.X or 53BP1 using immunofluorescence. The results obtained validate the reliability of our set-up in delivering equal radiation doses under all conditions employed. Although the initial numbers of γH2A.X and 53BP1 foci scored were similar under the different irradiation conditions, it was notable that the maximum foci level was reached at 60 min after irradiation with Bragg-peak protons, as compared to 30 min for plateau protons and photons. Interestingly, Bragg-peak protons induced larger and irregularly shaped γH2A.X and 53BP1 foci. Additionally, the resolution of these foci was delayed. These results suggest that Bragg-peak protons induce DNA damage of increased complexity which is difficult to process by the cellular repair apparatus.
Collapse
Affiliation(s)
- Sebastian Oeck
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany.
- Department of Therapeutic Radiology, Yale University School of Medicine, 15 York Street, New Haven, CT 06520, USA.
| | - Klaudia Szymonowicz
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany.
| | - Gesa Wiel
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany.
| | - Adam Krysztofiak
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany.
| | - Jamil Lambert
- West German Proton Therapy Centre Essen, University Hospital Essen, Am Muehlenbach 1, 45147 Essen, Germany.
| | - Benjamin Koska
- West German Proton Therapy Centre Essen, University Hospital Essen, Am Muehlenbach 1, 45147 Essen, Germany.
| | - George Iliakis
- Institute of Medical Radiation Biology; University of Duisburg-Essen; Medical School; Hufelandstr. 55, 45122 Essen, Germany.
| | - Beate Timmermann
- West German Proton Therapy Centre Essen, University Hospital Essen, Am Muehlenbach 1, 45147 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany.
| |
Collapse
|
29
|
Hlouschek J, Hansel C, Jendrossek V, Matschke J. The Mitochondrial Citrate Carrier (SLC25A1) Sustains Redox Homeostasis and Mitochondrial Metabolism Supporting Radioresistance of Cancer Cells With Tolerance to Cycling Severe Hypoxia. Front Oncol 2018; 8:170. [PMID: 29888201 PMCID: PMC5980958 DOI: 10.3389/fonc.2018.00170] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/01/2018] [Indexed: 12/29/2022] Open
Abstract
Pronounced resistance of lung cancer cells to radiotherapy and chemotherapy is a major barrier to successful treatment. Herein, both tumor hypoxia and the upregulation of the cellular antioxidant defense systems observed during malignant progression can contribute to radioresistance. We recently found that exposure to chronic cycling severe hypoxia/reoxygenation stress results in glutamine-dependent upregulation of cellular glutathione (GSH) levels and associated radiation resistance opening novel routes for tumor cell-specific radiosensitization. Here, we explored the role of the mitochondrial citrate carrier (SLC25A1) for the improved antioxidant defense of cancer cells with tolerance to acute and chronic severe hypoxia/reoxygenation stress and the use of pharmacologic SLC25A1 inhibition for tumor cell radiosensitization. Exposure to acute or chronic cycling severe hypoxia/reoxygenation stress triggered upregulated expression of SLC25A1 in lung cancer, prostate cancer, and glioblastoma cells in vitro. Interestingly, exposure to ionizing radiation (IR) further promoted SLC25A1 expression. Inhibition of SLC25A1 by 1,2,3-benzene-tricarboxylic acid (BTA) disturbed cellular and mitochondrial redox homeostasis, lowered mitochondrial metabolism, and reduced metabolic flexibility of cancer cells. Even more important, combining IR with BTA was able to overcome increased radioresistance induced by adaptation to chronic cycling severe hypoxia/reoxygenation stress. This radiosensitizing effect of BTA-treated cells was linked to increased reactive oxygen species and reduced DNA repair capacity. Of note, key findings could be reproduced when using the SLC25A1-inhibitor 4-Chloro-3-[[(3-nitrophenyl)amino]sulfonyl]-benzoic acid (CNASB). Moreover, in silico analysis of publically available databases applying the Kaplan–Meier plotter tool (kmplot.com) revealed that overexpression of SLC25A1 was associated with reduced survival of lung cancer patients suggesting a potential link to aggressive cancers. We show that SLC25A1 can contribute to the increased antioxidant defense of cancer cells allowing them to escape the cytotoxic effects of IR. Since upregulation of SLC25A1 is induced by adverse conditions in the tumor environment, exposure to IR, or both pharmacologic inhibition of SLC25A1 might be an effective strategy for radiosensitization of cancer cells particularly in chronically hypoxic tumor fractions.
Collapse
Affiliation(s)
- Julian Hlouschek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Hansel
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
30
|
Skowron MA, Melnikova M, van Roermund JGH, Romano A, Albers P, Thomale J, Schulz WA, Niegisch G, Hoffmann MJ. Multifaceted Mechanisms of Cisplatin Resistance in Long-Term Treated Urothelial Carcinoma Cell Lines. Int J Mol Sci 2018; 19:ijms19020590. [PMID: 29462944 PMCID: PMC5855812 DOI: 10.3390/ijms19020590] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 12/18/2022] Open
Abstract
Therapeutic efficacy of cisplatin-based treatment of late stage urothelial carcinoma (UC) is limited by chemoresistance. To elucidate underlying mechanisms and to develop new approaches for overcoming resistance, we generated long-term cisplatin treated (LTT) UC cell lines, characterised their cisplatin response, and determined the expression of molecules involved in cisplatin transport and detoxification, DNA repair, and apoptosis. Inhibitors of metallothioneins and Survivin were applied to investigate their ability to sensitise towards cisplatin. Cell growth, proliferation, and clonogenicity were examined after cisplatin treatment by MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, EdU (5-ethynyl-2’-deoxyuridine) incorporation assay, and Giemsa staining, respectively. Cell cycle distribution and apoptosis were quantified by flow cytometry. mRNA and protein expressions were measured by real-time quantitative (qRT)-PCR, western blot, or immunofluorescence staining. LTTs recovered rapidly from cisplatin stress compared to parental cells. In LTTs, to various extents, cisplatin exporters and metallothioneins were induced, cisplatin adduct levels and DNA damage were decreased, whereas expression of DNA repair factors and specific anti-apoptotic factors was elevated. Pharmacological inhibition of Survivin, but not of metallothioneins, sensitised LTTs to cisplatin, in an additive manner. LTTs minimise cisplatin-induced DNA damage and evade apoptosis by increased expression of anti-apoptotic factors. The observed diversity among the four LTTs highlights the complexity of cisplatin resistance mechanisms even within one tumour entity, explaining heterogeneity in patient responses to chemotherapy.
Collapse
Affiliation(s)
- Margaretha A Skowron
- Department of Urology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany.
| | - Margarita Melnikova
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen Medical School, 45122 Essen, Germany.
| | - Joep G H van Roermund
- Department of Urology, Maastricht University Medical Centre, 6202AZ Maastricht, The Netherlands.
| | - Andrea Romano
- Department of Obstetrics and Gynaecology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Peter Albers
- Department of Urology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany.
| | - Juergen Thomale
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen Medical School, 45122 Essen, Germany.
| | - Wolfgang A Schulz
- Department of Urology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany.
| | - Günter Niegisch
- Department of Urology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany.
| | - Michèle J Hoffmann
- Department of Urology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany.
| |
Collapse
|
31
|
Transcriptional repression of DNA repair genes is a hallmark and a cause of cellular senescence. Cell Death Dis 2018; 9:259. [PMID: 29449545 PMCID: PMC5833687 DOI: 10.1038/s41419-018-0300-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/19/2022]
Abstract
Cellular senescence response is (i) activated by numerous stresses, (ii) is characterized by a stable proliferation arrest, and (iii) by a set of specific features. Timely regulated senescence is thought to be beneficial, whereas chronic senescence such as during normal or premature aging is deleterious as it favors most, if not all, age-related diseases. In this study, using in-house or publicly available microarray analyses of transcriptomes of senescent cells, as well as analyses of the level of expression of several DNA repair genes by RT-qPCR and immunoblot, we show that repression of DNA repair gene expression is associated with cellular senescence. This repression is mediated by the RB/E2F pathway and it may play a causal role in senescence induction, as single DNA repair gene repression by siRNA induced features of premature senescence. Importantly, activating RB independently of direct DNA damage also results in repression of DNA repair genes and in the subsequent induction of DNA damage and senescence. The dogma is that DNA damage observed during cellular senescence is directly provoked by DNA lesions following genotoxic attack (UV, IR, and ROS) or by induction of replicative stress upon oncogenic activation. Our in vitro results support a largely unsuspected contribution of the loss of DNA repair gene expression in the induction and the accumulation of the DNA damage observed in most, if not all, kinds of cellular senescence, and thus in the induction of cellular senescence. Further demonstration using in vivo models will help to generalize our findings.
Collapse
|
32
|
Lim K, Chen E. Systematic Quantification of GFP-tagged Protein Foci in Schizosaccharomyces pombe Nuclei. Bio Protoc 2018; 8:e3117. [DOI: 10.21769/bioprotoc.3117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/12/2018] [Indexed: 11/02/2022] Open
|
33
|
Brill E, Yokoyama T, Nair J, Yu M, Ahn YR, Lee JM. Prexasertib, a cell cycle checkpoint kinases 1 and 2 inhibitor, increases in vitro toxicity of PARP inhibition by preventing Rad51 foci formation in BRCA wild type high-grade serous ovarian cancer. Oncotarget 2017; 8:111026-111040. [PMID: 29340034 PMCID: PMC5762302 DOI: 10.18632/oncotarget.22195] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/31/2017] [Indexed: 12/17/2022] Open
Abstract
PARP inhibitors (PARPi) have been effective in high-grade serous ovarian cancer (HGSOC), although clinical activity is limited against BRCA wild type HGSOC. The nearly universal loss of normal p53 regulation in HGSOCs causes dysfunction in the G1/S checkpoint, making tumor cells reliant on Chk1-mediated G2/M cell cycle arrest for DNA repair. Therefore, Chk1 is a reasonable target for a combination strategy with PARPi in treating BRCA wild type HGSOC. Here we investigated the combination of prexasertib mesylate monohydrate (LY2606368), a Chk1 and Chk2 inhibitor, and a PARP inhibitor, olaparib, in HGSOC cell lines (OVCAR3, OV90, PEO1 and PEO4) using clinically attainable concentrations. Our findings showed combination treatment synergistically decreased cell viability in all cell lines and induced greater DNA damage and apoptosis than the control and/or monotherapies (p<0.05). Treatment with olaparib in BRCA wild type HGSOC cells caused formation of Rad51 foci, whereas the combination treatment with prexasertib inhibited transnuclear localization of Rad51, a key protein in homologous recombination repair. Overall, our data provide evidence that prexasertib and olaparib combination resulted in synergistic cytotoxic effects against BRCA wild type HGSOC cells through reduced Rad51 foci formation and greater induction of apoptosis. This may be a novel therapeutic strategy for HGSOC.
Collapse
Affiliation(s)
- Ethan Brill
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Takuhei Yokoyama
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jayakumar Nair
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Minshu Yu
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yeong-Ran Ahn
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
34
|
Feng J, Lin J, Zhang P, Yang S, Sa Y, Feng Y. A novel automatic quantification method for high-content screening analysis of DNA double strand-break response. Sci Rep 2017; 7:9581. [PMID: 28852024 PMCID: PMC5574919 DOI: 10.1038/s41598-017-10063-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/02/2017] [Indexed: 01/09/2023] Open
Abstract
High-content screening is commonly used in studies of the DNA damage response. The double-strand break (DSB) is one of the most harmful types of DNA damage lesions. The conventional method used to quantify DSBs is γH2AX foci counting, which requires manual adjustment and preset parameters and is usually regarded as imprecise, time-consuming, poorly reproducible, and inaccurate. Therefore, a robust automatic alternative method is highly desired. In this manuscript, we present a new method for quantifying DSBs which involves automatic image cropping, automatic foci-segmentation and fluorescent intensity measurement. Furthermore, an additional function was added for standardizing the measurement of DSB response inhibition based on co-localization analysis. We tested the method with a well-known inhibitor of DSB response. The new method requires only one preset parameter, which effectively minimizes operator-dependent variations. Compared with conventional methods, the new method detected a higher percentage difference of foci formation between different cells, which can improve measurement accuracy. The effects of the inhibitor on DSB response were successfully quantified with the new method (p = 0.000). The advantages of this method in terms of reliability, automation and simplicity show its potential in quantitative fluorescence imaging studies and high-content screening for compounds and factors involved in DSB response.
Collapse
Affiliation(s)
- Jingwen Feng
- Department of Biomedical Engineering, Tianjin University, Tianjin, 300072, China
| | - Jie Lin
- Department of Biomedical Engineering, Tianjin University, Tianjin, 300072, China
| | - Pengquan Zhang
- Tianjin Optical Electrical Group Ltd, Tianjin, 300211, China
| | - Songnan Yang
- Tianjin Optical Electrical Group Ltd, Tianjin, 300211, China
| | - Yu Sa
- Department of Biomedical Engineering, Tianjin University, Tianjin, 300072, China.
| | - Yuanming Feng
- Department of Biomedical Engineering, Tianjin University, Tianjin, 300072, China. .,Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
35
|
Various Mechanisms Involve the Nuclear Factor (Erythroid-Derived 2)-Like (NRF2) to Achieve Cytoprotection in Long-Term Cisplatin-Treated Urothelial Carcinoma Cell Lines. Int J Mol Sci 2017; 18:ijms18081680. [PMID: 28767070 PMCID: PMC5578070 DOI: 10.3390/ijms18081680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/21/2017] [Accepted: 07/27/2017] [Indexed: 02/08/2023] Open
Abstract
Therapeutic efficacy of cisplatin-based chemotherapy for advanced-stage urothelial carcinoma (UC) is limited by drug resistance. The nuclear factor (erythroid-derived 2)-like 2 (NRF2) pathway is a major regulator of cytoprotective responses. We investigated its involvement in cisplatin resistance in long-term cisplatin treated UC cell lines (LTTs). Expression of NRF2 pathway components and targets was evaluated by qRT-PCR and western blotting in LTT sublines from four different parental cells. NRF2 transcriptional activity was determined by reporter assays and total glutathione (GSH) was quantified enzymatically. Effects of siRNA-mediated NRF2 knockdown on chemosensitivity were analysed by viability assays, γH2AX immunofluorescence, and flow cytometry. Increased expression of NRF2, its positive regulator p62/SQSTM1, and elevated NRF2 activity was observed in 3/4 LTTs, which correlated with KEAP1 expression. Expression of cytoprotective enzymes and GSH concentration were upregulated in some LTTs. NRF2 knockdown resulted in downregulation of cytoprotective enzymes and resensitised 3/4 LTTs towards cisplatin as demonstrated by reduced IC50 values, increased γH2AX foci formation, and elevated number of apoptotic cells. In conclusion, while LTT lines displayed diversity in NRF2 activation, NRF2 signalling contributed to cisplatin resistance in LTT lines, albeit in diverse ways. Accordingly, inhibition of NRF2 can be used to resensitise UC cells to cisplatin, but responses in patients may likewise be variable.
Collapse
|
36
|
Ishak CA, Coschi CH, Roes MV, Dick FA. Disruption of CDK-resistant chromatin association by pRB causes DNA damage, mitotic errors, and reduces Condensin II recruitment. Cell Cycle 2017; 16:1430-1439. [PMID: 28723239 DOI: 10.1080/15384101.2017.1338984] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Organization of chromatin structure is indispensible to the maintenance of genome integrity. The retinoblastoma tumor suppressor protein (pRB) mediates both transcriptional repression and chromatin organization, but the independent contributions of these functions have been difficult to study. Here, we utilize a synthetic Rb1 mutant allele (F832A) that maintains pRB association at cell cycle gene promoters, but disrupts a cyclin-dependent kinase (CDK)-resistant interaction with E2F1 to reduce occupancy of pRB on intergenic chromatin. Reduced pRB chromatin association increases spontaneous γH2AX deposition and aneuploidy. Our data indicates that the CDK-resistant pRB-E2F1 scaffold recruits Condensin II to major satellite repeats to stabilize chromatin structure in interphase and mitosis through mechanisms that are distinct from silencing of repetitive sequence expression.
Collapse
Affiliation(s)
- Charles A Ishak
- a London Regional Cancer Program , London , Ontario , Canada.,b Department of Biochemistry , Western University , London , Ontario , Canada
| | - Courtney H Coschi
- a London Regional Cancer Program , London , Ontario , Canada.,b Department of Biochemistry , Western University , London , Ontario , Canada
| | - Michael V Roes
- a London Regional Cancer Program , London , Ontario , Canada.,b Department of Biochemistry , Western University , London , Ontario , Canada
| | - Frederick A Dick
- a London Regional Cancer Program , London , Ontario , Canada.,b Department of Biochemistry , Western University , London , Ontario , Canada.,c Children's Health Research Institute , London , Ontario , Canada
| |
Collapse
|
37
|
Oeck S, Malewicz NM, Hurst S, Al-Refae K, Krysztofiak A, Jendrossek V. The Focinator v2-0 - Graphical Interface, Four Channels, Colocalization Analysis and Cell Phase Identification. Radiat Res 2017; 188:114-120. [PMID: 28492345 DOI: 10.1667/rr14746.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The quantitative analysis of foci plays an important role in various cell biological methods. In the fields of radiation biology and experimental oncology, the effect of ionizing radiation, chemotherapy or molecularly targeted drugs on DNA damage induction and repair is frequently performed by the analysis of protein clusters or phosphorylated proteins recruited to so called repair foci at DNA damage sites, involving for example γ-H2A.X, 53BP1 or RAD51. We recently developed "The Focinator" as a reliable and fast tool for automated quantitative and qualitative analysis of nuclei and DNA damage foci. The refined software is now even more user-friendly due to a graphical interface and further features. Thus, we included an R-script-based mode for automated image opening, file naming, progress monitoring and an error report. Consequently, the evaluation no longer required the attendance of the operator after initial parameter definition. Moreover, the Focinator v2-0 is now able to perform multi-channel analysis of four channels and evaluation of protein-protein colocalization by comparison of up to three foci channels. This enables for example the quantification of foci in cells of a specific cell cycle phase.
Collapse
Affiliation(s)
- Sebastian Oeck
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany
| | - Nathalie M Malewicz
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany
| | - Sebastian Hurst
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany
| | - Klaudia Al-Refae
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany
| | - Adam Krysztofiak
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, Virchowstrasse 173, 45122 Essen, Germany
| |
Collapse
|
38
|
Yokoyama T, Kohn EC, Brill E, Lee JM. Apoptosis is augmented in high-grade serous ovarian cancer by the combined inhibition of Bcl-2/Bcl-xL and PARP. Int J Oncol 2017; 50:1064-1074. [PMID: 28350129 PMCID: PMC5363883 DOI: 10.3892/ijo.2017.3914] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/17/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of our study was to evaluate possible synergistic cytotoxic effects of the combination treatment with the BH3-mimetic ABT-263 and the PARP inhibitor BMN 673 in high-grade serous ovarian cancer (HGSOC) cells using clinically achievable concentrations of each drug. In vitro cytotoxic effects of ABT-263 and BMN 673 were assessed by XTT assay in three HGSOC cell lines: OVCAR3, OVCAR8, and OV90 cells. Combination index values and synergy/antagonism volumes were used to determine synergy. The drug effects on DNA damage accumulation, cell cycle progression, apoptosis induction, and expression levels of Bcl-2 family proteins were examined to dissect molecular mechanisms. The combination treatment synergistically decreased cell viability in a concentration- and time-dependent manner in all cell lines; combination index values were <0.9 and synergy/antagonism volumes were >100 after 72 h of treatment. Clinically achievable concentrations of ABT-263 2 µM and BMN 673 25 nM were used to investigate mechanisms. No increase in γ-H2AX foci formation was observed with addition of ABT-263 to BMN 673 treatment. The combination treatment increased the sub-G1 and Annexin V-positive cell populations after 48 h compared with the control and each monotherapy. It also induced greater caspase-3/7 activity and PARP cleavage. ABT-263 alone and in combination with BMN 673 induced expression levels of Bim, a pro-apoptotic protein. In conclusion, the ABT-263 and BMN 673 combination resulted in synergistic cytotoxic effects against HGSOC cells through greater induction of apoptosis. This may be a novel therapeutic strategy for HGSOC.
Collapse
Affiliation(s)
- Takuhei Yokoyama
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Elise C. Kohn
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ethan Brill
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
39
|
Activating Akt1 mutations alter DNA double strand break repair and radiosensitivity. Sci Rep 2017; 7:42700. [PMID: 28209968 PMCID: PMC5314324 DOI: 10.1038/srep42700] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
The survival kinase Akt has clinical relevance to radioresistance. However, its contributions to the DNA damage response, DNA double strand break (DSB) repair and apoptosis remain poorly defined and often contradictory. We used a genetic approach to explore the consequences of genetic alterations of Akt1 for the cellular radiation response. While two activation-associated mutants with prominent nuclear access, the phospho-mimicking Akt1-TDSD and the clinically relevant PH-domain mutation Akt1-E17K, accelerated DSB repair and improved survival of irradiated Tramp-C1 murine prostate cancer cells and Akt1-knockout murine embryonic fibroblasts in vitro, the classical constitutively active membrane-targeted myrAkt1 mutant had the opposite effects. Interestingly, DNA-PKcs directly phosphorylated Akt1 at S473 in an in vitro kinase assay but not vice-versa. Pharmacological inhibition of DNA-PKcs or Akt restored radiosensitivity in tumour cells expressing Akt1-E17K or Akt1-TDSD. In conclusion, Akt1-mediated radioresistance depends on its activation state and nuclear localization and is accessible to pharmacologic inhibition.
Collapse
|
40
|
Lakatos P, Hegedűs C, Salazar Ayestarán N, Juarranz Á, Kövér KE, Szabó É, Virág L. The PARP inhibitor PJ-34 sensitizes cells to UVA-induced phototoxicity by a PARP independent mechanism. Mutat Res 2016; 790:31-40. [PMID: 27427773 DOI: 10.1016/j.mrfmmm.2016.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/28/2016] [Accepted: 07/04/2016] [Indexed: 12/24/2022]
Abstract
A combination of a photosensitizer with light of matching wavelength is a common treatment modality in various diseases including psoriasis, atopic dermatitis and tumors. DNA damage and production of reactive oxygen intermediates may impact pathological cellular functions and viability. Here we set out to investigate the role of the nuclear DNA nick sensor enzyme poly(ADP-ribose) polymerase 1 in photochemical treatment (PCT)-induced tumor cell killing. We found that silencing PARP-1 or inhibition of its enzymatic activity with Veliparib had no significant effect on the viability of A431 cells exposed to 8-methoxypsoralen (8-MOP) and UVA (2.5J/cm(2)) indicating that PARP-1 is not likely to be a key player in either cell survival or cell death of PCT-exposed cells. Interestingly, however, another commonly used PARP inhibitor PJ-34 proved to be a photosensitizer with potency equal to 8-MOP. Irradiation of PJ-34 with UVA caused changes both in the UV absorption and in the 1H NMR spectra of the compound with the latter suggesting UVA-induced formation of tautomeric forms of the compound. Characterization of the photosensitizing effect revealed that PJ-34+UVA triggers overproduction of reactive oxygen species, induces DNA damage, activation of caspase 3 and caspase 8 and internucleosomal DNA fragmentation. Cell death in this model could not be prevented by antioxidants (ascorbic acid, trolox, glutathione, gallotannin or cell permeable superoxide dismutase or catalase) but could be suppressed by inhibitors of caspase-3 and -8. In conclusion, PJ-34 is a photosensitizer and PJ-34+UVA causes DNA damage and caspase-mediated cell death independently of PARP-1 inhibition.
Collapse
Affiliation(s)
- Petra Lakatos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nerea Salazar Ayestarán
- Department of Biology, Faculty of Sciences, Universidad Autónoma of Madrid, 28049-Madrid, Spain
| | - Ángeles Juarranz
- Department of Biology, Faculty of Sciences, Universidad Autónoma of Madrid, 28049-Madrid, Spain
| | - Katalin E Kövér
- Department of Inorganic and Analytical Chemistry, Faculty of Sciences, University of Debrecen, Debrecen, Hungary
| | - Éva Szabó
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Debrecen, Hungary.
| |
Collapse
|