1
|
Llerena Schiffmacher DA, Pai YJ, Pines A, Vermeulen W. Transcription-coupled repair: tangled up in convoluted repair. FEBS J 2025. [PMID: 40272095 DOI: 10.1111/febs.70104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/08/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Significant progress has been made in understanding the mechanism of transcription-coupled nucleotide excision repair (TC-NER); however, numerous aspects remain elusive, including TC-NER regulation, lesion-specific and cell type-specific complex composition, structural insights, and lesion removal dynamics in living cells. This review summarizes and discusses recent advancements in TC-NER, focusing on newly identified interactors, mechanistic insights from cryo-electron microscopy (Cryo-EM) studies and live cell imaging, and the contribution of post-translational modifications (PTMs), such as ubiquitin, in regulating TC-NER. Furthermore, we elaborate on the consequences of TC-NER deficiencies and address the role of accumulated damage and persistent lesion-stalled RNA polymerase II (Pol II) as major drivers of the disease phenotype of Cockayne syndrome (CS) and its related disorders. In this context, we also discuss the severe effects of transcription-blocking lesions (TBLs) on neurons, highlighting their susceptibility to damage. Lastly, we explore the potential of investigating three-dimensional (3D) chromatin structure and phase separation to uncover further insights into this essential DNA repair pathway.
Collapse
Affiliation(s)
- Diana A Llerena Schiffmacher
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Yun Jin Pai
- Master Scientific Illustrations, Department of Anatomy and Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Alex Pines
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wim Vermeulen
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Zachayus A, Loup-Forest J, Cura V, Poterszman A. Nucleotide Excision Repair: Insights into Canonical and Emerging Functions of the Transcription/DNA Repair Factor TFIIH. Genes (Basel) 2025; 16:231. [PMID: 40004560 PMCID: PMC11855273 DOI: 10.3390/genes16020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Nucleotide excision repair (NER) is a universal cut-and-paste DNA repair mechanism that corrects bulky DNA lesions such as those caused by UV radiation, environmental mutagens, and some chemotherapy drugs. In this review, we focus on the human transcription/DNA repair factor TFIIH, a key player of the NER pathway in eukaryotes. This 10-subunit multiprotein complex notably verifies the presence of a lesion and opens the DNA around the damage via its XPB and XPD subunits, two proteins identified in patients suffering from Xeroderma Pigmentosum syndrome. Isolated as a class II gene transcription factor in the late 1980s, TFIIH is a prototypic molecular machine that plays an essential role in both DNA repair and transcription initiation and harbors a DNA helicase, a DNA translocase, and kinase activity. More recently, TFIIH subunits have been identified as participating in other cellular processes, including chromosome segregation during mitosis, maintenance of mitochondrial DNA integrity, and telomere replication.
Collapse
Affiliation(s)
- Amélie Zachayus
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Jules Loup-Forest
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Vincent Cura
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Arnaud Poterszman
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
3
|
Yavuz S, Abraham TE, Houtsmuller AB, van Royen ME. Phase Separation Mediated Sub-Nuclear Compartmentalization of Androgen Receptors. Cells 2024; 13:1693. [PMID: 39451211 PMCID: PMC11506798 DOI: 10.3390/cells13201693] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
The androgen receptor (AR), a member of the nuclear steroid hormone receptor family of transcription factors, plays a crucial role not only in the development of the male phenotype but also in the development and growth of prostate cancer. While AR structure and AR interactions with coregulators and chromatin have been studied in detail, improving our understanding of AR function in gene transcription regulation, the spatio-temporal organization and the role of microscopically discernible AR foci in the nucleus are still underexplored. This review delves into the molecular mechanisms underlying AR foci formation, focusing on liquid-liquid phase separation and its role in spatially organizing ARs and their binding partners within the nucleus at transcription sites, as well as the influence of 3D-genome organization on AR-mediated gene transcription.
Collapse
Affiliation(s)
- Selçuk Yavuz
- Department of Pathology, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.Y.); (M.E.v.R.)
| | - Tsion E. Abraham
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (T.E.A.)
| | - Adriaan B. Houtsmuller
- Department of Pathology, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.Y.); (M.E.v.R.)
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (T.E.A.)
| | - Martin E. van Royen
- Department of Pathology, Erasmus University Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.Y.); (M.E.v.R.)
| |
Collapse
|
4
|
Obermann R, Yemane B, Jarvis C, Franco FM, Kyriukha Y, Nolan W, Gohara B, Krezel AM, Wildman SA, Janetka JW. Small Molecule Antagonists of the DNA Repair ERCC1/XPA Protein-Protein Interaction. ChemMedChem 2024; 19:e202300648. [PMID: 38300970 PMCID: PMC11031295 DOI: 10.1002/cmdc.202300648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/03/2024]
Abstract
The DNA excision repair protein ERCC1 and the DNA damage sensor protein, XPA are highly overexpressed in patient samples of cisplatin-resistant solid tumors including lung, bladder, ovarian, and testicular cancer. The repair of cisplatin-DNA crosslinks is dependent upon nucleotide excision repair (NER) that is modulated by protein-protein binding interactions of ERCC1, the endonuclease, XPF, and XPA. Thus, inhibition of their function is a potential therapeutic strategy for the selective sensitization of tumors to DNA-damaging platinum-based cancer therapy. Here, we report on new small-molecule antagonists of the ERCC1/XPA protein-protein interaction (PPI) discovered using a high-throughput competitive fluorescence polarization binding assay. We discovered a unique structural class of thiopyridine-3-carbonitrile PPI antagonists that block a truncated XPA polypeptide from binding to ERCC1. Preliminary hit-to-lead studies from compound 1 reveal structure-activity relationships (SAR) and identify lead compound 27 o with an EC50 of 4.7 μM. Furthermore, chemical shift perturbation mapping by NMR confirms that 1 binds within the same site as the truncated XPA67-80 peptide. These novel ERCC1 antagonists are useful chemical biology tools for investigating DNA damage repair pathways and provide a good starting point for medicinal chemistry optimization as therapeutics for sensitizing tumors to DNA damaging agents and overcoming resistance to platinum-based chemotherapy.
Collapse
Affiliation(s)
| | | | - Cassie Jarvis
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Francisco M. Franco
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Yevhenii Kyriukha
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - William Nolan
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Beth Gohara
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Andrzej M. Krezel
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - Scott A. Wildman
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| | - James W. Janetka
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., Box 8231, St. Louis, MO 63110 USA
| |
Collapse
|
5
|
Yavuz S, Kabbech H, van Staalduinen J, Linder S, van Cappellen W, Nigg A, Abraham T, Slotman J, Quevedo M, Poot R, Zwart W, van Royen M, Grosveld F, Smal I, Houtsmuller A. Compartmentalization of androgen receptors at endogenous genes in living cells. Nucleic Acids Res 2023; 51:10992-11009. [PMID: 37791849 PMCID: PMC10639085 DOI: 10.1093/nar/gkad803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
A wide range of nuclear proteins are involved in the spatio-temporal organization of the genome through diverse biological processes such as gene transcription and DNA replication. Upon stimulation by testosterone and translocation to the nucleus, multiple androgen receptors (ARs) accumulate in microscopically discernable foci which are irregularly distributed in the nucleus. Here, we investigated the formation and physical nature of these foci, by combining novel fluorescent labeling techniques to visualize a defined chromatin locus of AR-regulated genes-PTPRN2 or BANP-simultaneously with either AR foci or individual AR molecules. Quantitative colocalization analysis showed evidence of AR foci formation induced by R1881 at both PTPRN2 and BANP loci. Furthermore, single-particle tracking (SPT) revealed three distinct subdiffusive fractional Brownian motion (fBm) states: immobilized ARs were observed near the labeled genes likely as a consequence of DNA-binding, while the intermediate confined state showed a similar spatial behavior but with larger displacements, suggesting compartmentalization by liquid-liquid phase separation (LLPS), while freely mobile ARs were diffusing in the nuclear environment. All together, we show for the first time in living cells the presence of AR-regulated genes in AR foci.
Collapse
Affiliation(s)
- Selçuk Yavuz
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hélène Kabbech
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jente van Staalduinen
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Simon Linder
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wiggert A van Cappellen
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Alex L Nigg
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tsion E Abraham
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Johan A Slotman
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marti Quevedo
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Raymond A Poot
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frank G Grosveld
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ihor Smal
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Adriaan B Houtsmuller
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Erasmus Optical Imaging Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Heemskerk T, van de Kamp G, Essers J, Kanaar R, Paul MW. Multi-scale cellular imaging of DNA double strand break repair. DNA Repair (Amst) 2023; 131:103570. [PMID: 37734176 DOI: 10.1016/j.dnarep.2023.103570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
Live-cell and high-resolution fluorescence microscopy are powerful tools to study the organization and dynamics of DNA double-strand break repair foci and specific repair proteins in single cells. This requires specific induction of DNA double-strand breaks and fluorescent markers to follow the DNA lesions in living cells. In this review, where we focused on mammalian cell studies, we discuss different methods to induce DNA double-strand breaks, how to visualize and quantify repair foci in living cells., We describe different (live-cell) imaging modalities that can reveal details of the DNA double-strand break repair process across multiple time and spatial scales. In addition, recent developments are discussed in super-resolution imaging and single-molecule tracking, and how these technologies can be applied to elucidate details on structural compositions or dynamics of DNA double-strand break repair.
Collapse
Affiliation(s)
- Tim Heemskerk
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gerarda van de Kamp
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiotherapy, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maarten W Paul
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
7
|
Llerena Schiffmacher DA, Kliza KW, Theil AF, Kremers GJ, Demmers JAA, Ogi T, Vermeulen M, Vermeulen W, Pines A. Live cell transcription-coupled nucleotide excision repair dynamics revisited. DNA Repair (Amst) 2023; 130:103566. [PMID: 37716192 DOI: 10.1016/j.dnarep.2023.103566] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 09/18/2023]
Abstract
Transcription-blocking lesions are specifically targeted by transcription-coupled nucleotide excision repair (TC-NER), which prevents DNA damage-induced cellular toxicity and maintains proper transcriptional processes. TC-NER is initiated by the stalling of RNA polymerase II (RNAPII), which triggers the assembly of TC-NER-specific proteins, namely CSB, CSA and UVSSA, which collectively control and drive TC-NER progression. Previous research has revealed molecular functions for these proteins, however, exact mechanisms governing the initiation and regulation of TC-NER, particularly at low UV doses have remained elusive, partly due to technical constraints. In this study, we employ knock-in cell lines designed to target the endogenous CSB gene locus with mClover, a GFP variant. Through live cell imaging, we uncover the intricate molecular dynamics of CSB in response to physiologically relevant UV doses. We showed that the DNA damage-induced association of CSB with chromatin is tightly regulated by the CSA-containing ubiquitin-ligase CRL complex (CRL4CSA). Combining the CSB-mClover knock-in cell line with SILAC-based GFP-mediated complex isolation and mass-spectrometry-based proteomics, revealed novel putative CSB interactors as well as discernible variations in complex composition during distinct stages of TC-NER progression. Our work not only provides molecular insight into TC-NER, but also illustrates the versatility of endogenously tagging fluorescent and affinity tags.
Collapse
Affiliation(s)
- Diana A Llerena Schiffmacher
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr Molewaterplein 40, Rotterdam 3015 GD, the Netherlands
| | - Katarzyna W Kliza
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Oncode Institute, Radboud University Nijmegen, Geert Grooteplein Zuid 28, Nijmegen 6525 GA, the Netherlands
| | - Arjan F Theil
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr Molewaterplein 40, Rotterdam 3015 GD, the Netherlands
| | - Gert-Jan Kremers
- Optical Imaging Centre, Erasmus University Medical Center, Dr Molewaterplein 40, Rotterdam 3015 GD, the Netherlands
| | - Jeroen A A Demmers
- Proteomics Center, Erasmus University Medical Center, Dr Molewaterplein 40, Rotterdam 3015 GD, the Netherland
| | - Tomoo Ogi
- Department of Human Genetics and Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan; Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Oncode Institute, Radboud University Nijmegen, Geert Grooteplein Zuid 28, Nijmegen 6525 GA, the Netherlands; Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands
| | - Wim Vermeulen
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr Molewaterplein 40, Rotterdam 3015 GD, the Netherlands.
| | - Alex Pines
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr Molewaterplein 40, Rotterdam 3015 GD, the Netherlands.
| |
Collapse
|
8
|
Forgie BN, Prakash R, Telleria CM. Revisiting the Anti-Cancer Toxicity of Clinically Approved Platinating Derivatives. Int J Mol Sci 2022; 23:15410. [PMID: 36499737 PMCID: PMC9793759 DOI: 10.3390/ijms232315410] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Cisplatin (CDDP), carboplatin (CP), and oxaliplatin (OXP) are three platinating agents clinically approved worldwide for use against a variety of cancers. They are canonically known as DNA damage inducers; however, that is only one of their mechanisms of cytotoxicity. CDDP mediates its effects through DNA damage-induced transcription inhibition and apoptotic signalling. In addition, CDDP targets the endoplasmic reticulum (ER) to induce ER stress, the mitochondria via mitochondrial DNA damage leading to ROS production, and the plasma membrane and cytoskeletal components. CP acts in a similar fashion to CDDP by inducing DNA damage, mitochondrial damage, and ER stress. Additionally, CP is also able to upregulate micro-RNA activity, enhancing intrinsic apoptosis. OXP, on the other hand, at first induces damage to all the same targets as CDDP and CP, yet it is also capable of inducing immunogenic cell death via ER stress and can decrease ribosome biogenesis through its nucleolar effects. In this comprehensive review, we provide detailed mechanisms of action for the three platinating agents, going beyond their nuclear effects to include their cytoplasmic impact within cancer cells. In addition, we cover their current clinical use and limitations, including side effects and mechanisms of resistance.
Collapse
Affiliation(s)
- Benjamin N. Forgie
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Rewati Prakash
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
9
|
Wang G, Heij LR, Liu D, Dahl E, LANG SA, Ulmer TF, LUEDDE T, Neumann UP, Bednarsch J. The Role of Single-Nucleotide Polymorphisms in Cholangiocarcinoma: A Systematic Review. Cancers (Basel) 2022; 14:cancers14235969. [PMID: 36497451 PMCID: PMC9739277 DOI: 10.3390/cancers14235969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Single-nucleotide polymorphisms (SNPs) play an essential role in various malignancies, but their role in cholangiocarcinoma (CCA) remains to be elucidated. Therefore, the purpose of this systematic review was to evaluate the association between SNPs and CCA, focusing on tumorigenesis and prognosis. A systematic literature search was carried out using PubMed, Embase, Web of Science and the Cochrane database for the association between SNPs and CCA, including literature published between January 2000 and April 2022. This systematic review compiles 43 SNPs in 32 genes associated with CCA risk, metastatic progression and overall prognosis based on 34 studies. Susceptibility to CCA was associated with SNPs in genes related to inflammation (PTGS2/COX2, IL6, IFNG/IFN-γ, TNF/TNF-α), DNA repair (ERCC1, MTHFR, MUTYH, XRCC1, OGG1), detoxification (NAT1, NAT2 and ABCC2), enzymes (SERPINA1, GSTO1, APOBEC3A, APOBEC3B), RNA (HOTAIR) and membrane-based proteins (EGFR, GAB1, KLRK1/NKG2D). Overall oncological prognosis was also related to SNPs in eight genes (GNB3, NFE2L2/NRF2, GALNT14, EGFR, XRCC1, EZH2, GNAS, CXCR1). Our findings indicate that multiple SNPs play different roles at various stages of CCA and might serve as biomarkers guiding treatment and allowing oncological risk assessment. Considering the differences in SNP detection methods, patient ethnicity and corresponding environmental factors, more large-scale multicentric investigations are needed to fully determine the potential of SNP analysis for CCA susceptibility prediction and prognostication.
Collapse
Affiliation(s)
- Guanwu Wang
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Lara Rosaline Heij
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6211 LK Maastricht, The Netherlands
- Department of Pathology, Erasmus Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Dong Liu
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Sven Arke LANG
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Tom Florian Ulmer
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Tom LUEDDE
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Ulf Peter Neumann
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Department of Surgery, Maastricht University Medical Center (MUMC), 6229 HX Maastricht, The Netherlands
| | - Jan Bednarsch
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
10
|
Vascular Ageing Features Caused by Selective DNA Damage in Smooth Muscle Cell. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2308317. [PMID: 34504640 PMCID: PMC8423575 DOI: 10.1155/2021/2308317] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022]
Abstract
Persistently unrepaired DNA damage has been identified as a causative factor for vascular ageing. We have previously shown that a defect in the function or expression of the DNA repair endonuclease ERCC1 (excision repair cross complement 1) in mice leads to accelerated, nonatherosclerotic ageing of the vascular system from as early as 8 weeks after birth. Removal of ERCC1 from endothelial alone partly explains this ageing, as shown in endothelial-specific Ercc1 knockout mice. In this study, we determined vascular ageing due to DNA damage in vascular smooth muscle cells, as achieved by smooth muscle-selective genetic removal of ERCC1 DNA repair in mice (SMC-KO: SM22αCre+ Ercc1fl/-). Vascular ageing features in SMC-KO and their wild-type littermates (WT: SM22αCre+ Ercc1fl/+) were examined at the age of 14 weeks and 25 weeks. Both SMC-KO and WT mice were normotensive. Compared to WT, SMC-KO showed a reduced heart rate, fractional shortening, and cardiac output. SMC-KO showed progressive features of nonatherosclerotic vascular ageing as they aged from 14 to 25 weeks. Decreased subcutaneous microvascular dilatation and increased carotid artery stiffness were observed. Vasodilator responses measured in aortic rings in organ baths showed decreased endothelium-dependent and endothelium-independent responses, mostly due to decreased NO-cGMP signaling. NADPH oxidase 2 and phosphodiesterase 1 inhibition improved dilations. SMC-KO mice showed elevated levels of various cytokines that indicate a balance shift in pro- and anti-inflammatory pathways. In conclusion, SMC-KO mice showed a progressive vascular ageing phenotype in resistant and conduit arteries that is associated with cardiac remodeling and contractile dysfunction. The changes induced by DNA damage might be limited to VSMC but eventually affect EC-mediated responses. The fact that NADPH oxidase 2 as wells as phosphodiesterase 1 inhibition restores vasodilation suggests that both decreased NO bioavailability and cGMP degradation play a role in local vascular smooth muscle cell ageing induced by DNA damage.
Collapse
|
11
|
Wong CT, Oh DH. Vitamin D Receptor Promotes Global Nucleotide Excision Repair by Facilitating XPC Dissociation from Damaged DNA. J Invest Dermatol 2021; 141:1656-1663. [PMID: 33524369 DOI: 10.1016/j.jid.2020.11.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022]
Abstract
Vitamin D receptor (VDR) is important for normal DNA repair, although the mechanism by which it acts is unclear. After focal UV irradiation to create subnuclear spots of DNA damage, epidermal keratinocytes from VDR-null mice as well as human epidermal keratinocytes depleted of VDR with small interfering RNA removed pyrimidine-pyrimidone (6-4) photoproducts more slowly than control cells. Costaining with antibodies to XPC, the DNA damage recognition sensor that initiates nucleotide excision repair, showed that XPC rapidly accumulated at spots of damage and gradually faded in control human keratinocytes. In VDR-depleted keratinocytes, XPC associated with DNA damage with comparable efficiency; however, XPC's dissociation dynamics were altered so that significantly more XPC was bound and retained over time than in control cells. The XPF endonuclease, which acts subsequently in nucleotide excision repair, bound and dissociated with comparable kinetics in control and VDR-depleted cells, but the extent of binding was reduced in the latter. These results as well as kinetic modeling of the data suggest that VDR's importance in the repair of UV-induced DNA damage is mediated in part by its ability to facilitate the dissociation of XPC from damaged DNA for the normal recruitment and assembly of other repair proteins to proceed.
Collapse
Affiliation(s)
- Christian T Wong
- Dermatology Research Unit, San Francisco VA Health Care System, San Francisco, California, USA; Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Dennis H Oh
- Dermatology Research Unit, San Francisco VA Health Care System, San Francisco, California, USA; Department of Dermatology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
12
|
Parameter estimation in fluorescence recovery after photobleaching: quantitative analysis of protein binding reactions and diffusion. J Math Biol 2021; 83:1. [PMID: 34129100 PMCID: PMC8205911 DOI: 10.1007/s00285-021-01616-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 09/15/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Fluorescence recovery after photobleaching (FRAP) is a common experimental method for investigating rates of molecular redistribution in biological systems. Many mathematical models of FRAP have been developed, the purpose of which is usually the estimation of certain biological parameters such as the diffusivity and chemical reaction rates of a protein, this being accomplished by fitting the model to experimental data. In this article, we consider a two species reaction–diffusion FRAP model. Using asymptotic analysis, we derive new FRAP recovery curve approximation formulae, and formally re-derive existing ones. On the basis of these formulae, invoking the concept of Fisher information, we predict, in terms of biological and experimental parameters, sufficient conditions to ensure that the values all model parameters can be estimated from data. We verify our predictions with extensive computational simulations. We also use computational methods to investigate cases in which some or all biological parameters are theoretically inestimable. In these cases, we propose methods which can be used to extract the maximum possible amount of information from the FRAP data.
Collapse
|
13
|
Apelt K, White SM, Kim HS, Yeo JE, Kragten A, Wondergem AP, Rooimans MA, González-Prieto R, Wiegant WW, Lunke S, Flanagan D, Pantaleo S, Quinlan C, Hardikar W, van Attikum H, Vertegaal AC, Wilson BT, Wolthuis RM, Schärer OD, Luijsterburg MS. ERCC1 mutations impede DNA damage repair and cause liver and kidney dysfunction in patients. J Exp Med 2021; 218:e20200622. [PMID: 33315086 PMCID: PMC7927433 DOI: 10.1084/jem.20200622] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/25/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
ERCC1-XPF is a multifunctional endonuclease involved in nucleotide excision repair (NER), interstrand cross-link (ICL) repair, and DNA double-strand break (DSB) repair. Only two patients with bi-allelic ERCC1 mutations have been reported, both of whom had features of Cockayne syndrome and died in infancy. Here, we describe two siblings with bi-allelic ERCC1 mutations in their teenage years. Genomic sequencing identified a deletion and a missense variant (R156W) within ERCC1 that disrupts a salt bridge below the XPA-binding pocket. Patient-derived fibroblasts and knock-in epithelial cells carrying the R156W substitution show dramatically reduced protein levels of ERCC1 and XPF. Moreover, mutant ERCC1 weakly interacts with NER and ICL repair proteins, resulting in diminished recruitment to DNA damage. Consequently, patient cells show strongly reduced NER activity and increased chromosome breakage induced by DNA cross-linkers, while DSB repair was relatively normal. We report a new case of ERCC1 deficiency that severely affects NER and considerably impacts ICL repair, which together result in a unique phenotype combining short stature, photosensitivity, and progressive liver and kidney dysfunction.
Collapse
Affiliation(s)
- Katja Apelt
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Susan M. White
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Hyun Suk Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Jung-Eun Yeo
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Angela Kragten
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Martin A. Rooimans
- Section of Oncogenetics, Department of Clinical Genetics, Vrije Universiteit Medical Center and Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Román González-Prieto
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Wouter W. Wiegant
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastian Lunke
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Parkville, Australia
- Department of Pathology, University of Melbourne, Parkville, Australia
| | - Daniel Flanagan
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Parkville, Australia
| | - Sarah Pantaleo
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Parkville, Australia
| | - Catherine Quinlan
- Department of Paediatrics, University of Melbourne, Parkville, Australia
- Department of Nephrology, Royal Children’s Hospital, Melbourne, Australia
- Department of Kidney Regeneration, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Winita Hardikar
- Department of Paediatrics, University of Melbourne, Parkville, Australia
- Department of Gastroenterology, Royal Children's Hospital, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Parkville, Australia
| | - Haico van Attikum
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Alfred C.O. Vertegaal
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Brian T. Wilson
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK
- Northern Genetics Service, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, International Centre for Life, Newcastle upon Tyne, UK
- Department of Clinical Genetics, Great Ormond Street Hospital, London, UK
| | - Rob M.F. Wolthuis
- Section of Oncogenetics, Department of Clinical Genetics, Vrije Universiteit Medical Center and Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Orlando D. Schärer
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | | |
Collapse
|
14
|
Tissue-Specific DNA Repair Activity of ERCC-1/XPF-1. Cell Rep 2021; 34:108608. [PMID: 33440146 DOI: 10.1016/j.celrep.2020.108608] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 01/14/2023] Open
Abstract
Hereditary DNA repair defects affect tissues differently, suggesting that in vivo cells respond differently to DNA damage. Knowledge of the DNA damage response, however, is largely based on in vitro and cell culture studies, and it is currently unclear whether DNA repair changes depending on the cell type. Here, we use in vivo imaging of the nucleotide excision repair (NER) endonuclease ERCC-1/XPF-1 in C. elegans to demonstrate tissue-specific NER activity. In oocytes, XPF-1 functions as part of global genome NER (GG-NER) to ensure extremely rapid removal of DNA-helix-distorting lesions throughout the genome. In contrast, in post-mitotic neurons and muscles, XPF-1 participates in NER of transcribed genes only. Strikingly, muscle cells appear more resistant to the effects of DNA damage than neurons. These results suggest a tissue-specific organization of the DNA damage response and may help to better understand pleiotropic and tissue-specific consequences of accumulating DNA damage.
Collapse
|
15
|
Robu M, Shah RG, Shah GM. Methods to Study Intracellular Movement and Localization of the Nucleotide Excision Repair Proteins at the DNA Lesions in Mammalian Cells. Front Cell Dev Biol 2020; 8:590242. [PMID: 33282869 PMCID: PMC7705073 DOI: 10.3389/fcell.2020.590242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/27/2020] [Indexed: 11/13/2022] Open
Abstract
Nucleotide excision repair (NER) is the most versatile DNA repair pathway that removes a wide variety of DNA lesions caused by different types of physical and chemical agents, such as ultraviolet radiation (UV), environmental carcinogen benzo[a]pyrene and anti-cancer drug carboplatin. The mammalian NER utilizes more than 30 proteins, in a multi-step process that begins with the lesion recognition within seconds of DNA damage to completion of repair after few hours to several days. The core proteins and their biochemical reactions are known from in vitro DNA repair assays using purified proteins, but challenge was to understand the dynamics of their rapid recruitment and departure from the lesion site and their coordination with other proteins and post-translational modifications to execute the sequential steps of repair. Here, we provide a brief overview of various techniques developed by different groups over last 20 years to overcome these challenges. However, more work is needed for a comprehensive knowledge of all aspects of mammalian NER. With this aim, here we provide detailed protocols of three simple yet innovative methods developed by many teams that range from local UVC irradiation to in situ extraction and sub-cellular fractionation that will permit study of endogenous as well as exogenous NER proteins in any cellular model. These methods do not require unique reagents or specialized instruments, and will allow many more laboratories to explore this repair pathway in different models. These techniques would reveal intracellular movement of these proteins to the DNA lesion site, their interactions with other proteins during repair and the effect of post-translational modifications on their functions. We also describe how these methods led us to identify hitherto unexpected role of poly(ADP-ribose) polymerase-1 (PARP1) in NER. Collectively these three simple techniques can provide an initial assessment of the functions of known and unknown proteins in the core or auxiliary events associated with mammalian NER. The results from these techniques could serve as a solid foundation and a justification for more detailed studies in NER using specialized reagents and more sophisticated tools. They can also be suitably modified to study other cellular processes beyond DNA repair.
Collapse
Affiliation(s)
- Mihaela Robu
- CHU de Québec Université Laval Research Centre (site CHUL), Laboratory for Skin Cancer Research and Axe Neuroscience, Québec, QC, Canada
| | - Rashmi G Shah
- CHU de Québec Université Laval Research Centre (site CHUL), Laboratory for Skin Cancer Research and Axe Neuroscience, Québec, QC, Canada
| | - Girish M Shah
- CHU de Québec Université Laval Research Centre (site CHUL), Laboratory for Skin Cancer Research and Axe Neuroscience, Québec, QC, Canada
| |
Collapse
|
16
|
Li H, Zhou L, Ma J, Zhu Y, Fan J, Li N, Zheng Y, Sha T, Zhai Z, Ma B, Dai Z. Distribution and susceptibility of ERCC1/XPF gene polymorphisms in Han and Uygur women with breast cancer in Xinjiang, China. Cancer Med 2020; 9:9571-9580. [PMID: 33067872 PMCID: PMC7774751 DOI: 10.1002/cam4.3547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/21/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022] Open
Abstract
This study aimed to explore the roles of ERCC1/XPF gene polymorphisms in the occurrence of breast cancer in the Uygur and Han ethnic groups in Xinjiang, China. Single nucleotide polymorphisms (SNPs) were detected by TaqMan real-time PCR. The rs11615 G>A and rs2276466 C>G variant frequencies were higher in Uygur patients with breast cancer than in Han patients, while the frequency of rs2298881 C>A was higher in Han patients. We found that rs2298881 C>A (CA vs. CC: OR = 0.35, 95% CI = 0.20-0.60; AA vs. CC: OR = 0.13, 95% CI = 0.04-0.34; CA + AA vs. CC: OR = 0.33, 95% CI = 0.18-0.51; AA vs. CA + CC: OR = 0.24, 95% CI = 0.08-0.62; CA vs. AA + CC: OR = 0.49, 95% CI = 0.29-0.82) was associated with a reduced breast cancer risk and rs3212986 C>A (AA vs. CC: OR = 4.80, 95% CI = 1.79-15.29,; CA+AA vs. CC: OR = 1.71, 95% CI = 1.06-2.77; AA vs. CA+CC: OR = 4.12, 95% CI =1.58-12.89) and rs11615 G > A (AA vs. GG: OR = 3.49, 95% CI =1.54-8.55; GA + AA vs. GG: OR = 1.98, 95% CI = 1.21-3.27; AA vs. GA+GG: OR = 2.87, 95% CI = 1.30-6.85) were associated with an elevated breast cancer risk among Uygur individuals. In addition, Uygur patients with breast cancer with 2-3 combined risk genotypes of ERCC1 had a higher risk than patients with 0-1 risk genotypes (OR = 2.91; 95% CI = 1.54-5.71, p = 0.001). However, we failed to detect a statistically significant association between ERCC1/XPF polymorphisms and breast cancer risk in five genetic models among Han individuals. Our results showed that ERCC1/XPF gene polymorphisms predispose Uygur individuals to breast cancer; this finding should be verified by further large-scale analyses.
Collapse
Affiliation(s)
- Hongtao Li
- Department of Breast Head and Neck SurgeryThe 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor HospitalUrumqiChina
| | - Linghui Zhou
- Department of Breast SurgeryThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Jing Ma
- Department of Breast Head and Neck SurgeryThe 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor HospitalUrumqiChina
| | - Yuyao Zhu
- Department of OncologyThe Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Jingjing Fan
- Department of Breast Head and Neck SurgeryThe 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor HospitalUrumqiChina
| | - Na Li
- Department of OncologyThe Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Yi Zheng
- Department of OncologyThe Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Tong Sha
- Department of Breast Head and Neck SurgeryThe 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor HospitalUrumqiChina
| | - Zhen Zhai
- Department of OncologyThe Second Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Binlin Ma
- Department of Breast Head and Neck SurgeryThe 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor HospitalUrumqiChina
| | - Zhijun Dai
- Department of Breast SurgeryThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
17
|
Abstract
The nucleolus is the most prominent nuclear body and serves a fundamentally important biological role as a site of ribonucleoprotein particle assembly, primarily dedicated to ribosome biogenesis. Despite being one of the first intracellular structures visualized historically, the biophysical rules governing its assembly and function are only starting to become clear. Recent studies have provided increasing support for the concept that the nucleolus represents a multilayered biomolecular condensate, whose formation by liquid-liquid phase separation (LLPS) facilitates the initial steps of ribosome biogenesis and other functions. Here, we review these biophysical insights in the context of the molecular and cell biology of the nucleolus. We discuss how nucleolar function is linked to its organization as a multiphase condensate and how dysregulation of this organization could provide insights into still poorly understood aspects of nucleolus-associated diseases, including cancer, ribosomopathies and neurodegeneration as well as ageing. We suggest that the LLPS model provides the starting point for a unifying quantitative framework for the assembly, structural maintenance and function of the nucleolus, with implications for gene regulation and ribonucleoprotein particle assembly throughout the nucleus. The LLPS concept is also likely useful in designing new therapeutic strategies to target nucleolar dysfunction.
Collapse
|
18
|
Liao X, Li Y, Li H, Huang W, Wang H, Xie W. Expression and Clinical Significance of ERCC1 and XPF in Human Hepatocellular Carcinoma. Onco Targets Ther 2020; 13:1059-1072. [PMID: 32099408 PMCID: PMC7007798 DOI: 10.2147/ott.s237916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/20/2020] [Indexed: 12/22/2022] Open
Abstract
Objective To investigate the correlation between the ERCC1 and XPF expression and the clinicopathological parameters of hepatocellular carcinoma (HCC) patients through assessment of the expression of the DNA repair genes ERCC1 and XPF. Methods ERCC1 and XPF mRNA expression in HCC (n= 177) and adjacent para-cancer tissues (n=21) were assessed by RT-PCR. The correlation between ERCC1 and XPF expression, clinicopathological features and HCC prognosis were compared. Results ERCC1 expression in liver cancer tissues was significantly lower than that of adjacent tissues (9.5% (2/21) vs 38.1% (8/21); P<0.05). The positive expression rates of XPF in liver cancer tissues was lower than that of adjacent tissues (14.3% (3/21) vs 71.4% (15/21); P<0.05). ERCC1 and XPF expression were associated with hepatic capsular invasion and microvascular invasion. HCC patients with hepatic capsular invasion and microvascular tumor embolus formation had significantly lower levels of ERCC1 and XPF mRNA than those without hepatic capsular invasion and microvascular tumor embolus formation (P<0.05). In addition, ERCC1 expression was associated with TNM staging of HCC. The expression of ERCC1 mRNA in patients with stage II and III HCC were lower than that of patients with stage I HCC (P<0.05). The low levels of ERCC1 and XPF mRNA significantly correlated with relapse-free survival times (RFS) in HCC patients. The median RFS of the low ERCC1 expression group and low XPF expression group were shorter than those of the high expression group (15.0 months vs 32.0 months, P<0.05) and (19.0 months vs 33.0 months, P<0.05). The decrease in XPF mRNA expression was significantly associated with the overall survival (OS) of HCC patients. The median OS in the low XPF expression group was shorter than that of the high expression group (46.0 months vs 78.0 months, P<0.05). However, no significant difference in OS between the low ERCC1 expression group and the high ERCC1 expression groups were observed (63.0 months vs 64.0 months, P>0.05). Multivariate analysis showed that tumor size and the extent of differentiation were independent factors affecting the RFS in HCC patients (P<0.05). The extent of differentiation and XPF were independent factors affecting the OS in HCC (P<0.05). Conclusion The expression in ERCC1 and XPF were low in HCC and associated with early relapse after HCC surgery. Low XPF expression may be a potential indicator of a high risk of death.
Collapse
Affiliation(s)
- Xiaoli Liao
- Department of First Chemotherapy, Affiliated Guangxi Medical University Cancer Hospital, Nanning 530021, People's Republic of China
| | - Yongqiang Li
- Department of First Chemotherapy, Affiliated Guangxi Medical University Cancer Hospital, Nanning 530021, People's Republic of China
| | - Hualan Li
- Department of First Chemotherapy, Affiliated Guangxi Medical University Cancer Hospital, Nanning 530021, People's Republic of China
| | - Wenfeng Huang
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, People's Republic of China
| | - Hongxue Wang
- Department of Fifth Chemotherapy, Affiliated Guangxi Medical University Cancer Hospital, Nanning 530021, People's Republic of China
| | - Weimin Xie
- Department of Fifth Chemotherapy, Affiliated Guangxi Medical University Cancer Hospital, Nanning 530021, People's Republic of China
| |
Collapse
|
19
|
Ten Broeke T, Honing H, Brandsma AM, Jacobino S, Bakema JE, Kanters D, van der Linden JAM, Bracke M, Koenderman L, Leusen JHW. FcαRI Dynamics Are Regulated by GSK-3 and PKCζ During Cytokine Mediated Inside-Out Signaling. Front Immunol 2019; 9:3191. [PMID: 30766540 PMCID: PMC6365424 DOI: 10.3389/fimmu.2018.03191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 12/31/2018] [Indexed: 12/03/2022] Open
Abstract
IgA binding to FcαRI (CD89) is rapidly enhanced by cytokine induced inside-out signaling. Dephosphorylation of serine 263 in the intracellular tail of FcαRI by PP2A and PI3K activation are instrumental in this process. To further investigate these signaling pathways, we targeted downstream kinases of PI3K. Our experiments revealed that PI3K activates PKCζ, which subsequently inhibits GSK-3, a constitutively active kinase in resting cells and found here to be associated with FcαRI. We propose that GSK-3 maintains FcαRI in an inactive state at homeostatic conditions. Upon cytokine stimulation, GSK-3 is inactivated through a PI3K-PKCζ pathway, preventing the maintenance of phosphorylated inactive FcαRI. The concomitantly activated PP2A is then able to dephosphorylate and activate FcαRI. Moreover, FRAP and FLIP studies showed that FcαRI activation coincides with an increased mobile fraction of the receptor. This can enhance FcαRI valency and contribute to stronger avidity for IgA immune complexes. This tightly regulated inside-out signaling pathway allows leukocytes to respond rapidly and efficiently to their environment and could be exploited to enhance the efficacy of future IgA therapeutics.
Collapse
Affiliation(s)
- Toine Ten Broeke
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| | - Henk Honing
- Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Arianne M Brandsma
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| | - Shamir Jacobino
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| | - Jantine E Bakema
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Tumor Biology Section, Department of Otolaryngology, Head-Neck Surgery, VU University Medical Center, Amsterdam, Netherlands
| | - Deon Kanters
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Jan A M van der Linden
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Madelon Bracke
- Department of Pharmacoepidemiology and Pharmacotherapy, University Medical Center, Utrecht, Netherlands
| | - Leo Koenderman
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center, Utrecht, Netherlands
| | - Jeanette H W Leusen
- Laboratory of Translational Immunology, University Medical Center, Utrecht, Netherlands
| |
Collapse
|
20
|
Leow JJ, Bedke J, Chamie K, Collins JW, Daneshmand S, Grivas P, Heidenreich A, Messing EM, Royce TJ, Sankin AI, Schoenberg MP, Shipley WU, Villers A, Efstathiou JA, Bellmunt J, Stenzl A. SIU–ICUD consultation on bladder cancer: treatment of muscle-invasive bladder cancer. World J Urol 2019; 37:61-83. [DOI: 10.1007/s00345-018-2606-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/12/2018] [Indexed: 01/09/2023] Open
|
21
|
Sharma K, Darvas M, Keene CD, Niedernhofer LJ, Ladiges W. Modeling Alzheimer's disease in progeria mice. An age-related concept. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2018; 8:1524815. [PMID: 30319737 PMCID: PMC6179061 DOI: 10.1080/20010001.2018.1524815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The prevalence of Alzheimer’s disease (AD) is expected to dramatically increase in older people worldwide. Efforts to find disease-modifying treatments have been largely unsuccessful because of the focus on disease-specific pathogenesis, and lack of animal models to study AD in the context of aging and age-related co-morbidities. The geroscience approach to studying AD would suggest that modulation of aging per se would be a useful strategy, but a mammalian model system that combines both aging and AD is not available. One approach to study old age and AD is to utilize murine models of progeroid syndrome, which can provide a number of advantages not only for basic aging biology but also for preclinical drug testing. A progeria background, such as the Ercc1 mutant mouse (Ercc1−/Δ), provides an aging component not seen in current murine models of AD that lack age-related co-morbidities typical of AD patients. Ercc1−/Δ mice experience the same types of stochastic endogenous DNA damage as WT mice, but accumulate lesions faster due to impaired DNA repair, which accelerates the normal aging process by 6-fold. These mice do not show frank AD pathology but represent a predisposed or hypersensitive environment for AD pathology, where pathogenic elements of AD can be introduced, either by crossing with well-established AD transgenic mouse lines, or transcranial stereotaxic delivery directly into the brain. Since Ercc1−/Δ mice age five to six times faster than WT mice, very rapid characterization and testing of therapeutic interventions is possible. Studies are urgently needed to capitalize on the highly informative potential of this novel AD mouse model.
Collapse
Affiliation(s)
- Kavita Sharma
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Martin Darvas
- Department of Pathology, Division of Neuropathology, School of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Department of Pathology, Division of Neuropathology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Warren Ladiges
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
22
|
Sabatella M, Theil AF, Ribeiro-Silva C, Slyskova J, Thijssen K, Voskamp C, Lans H, Vermeulen W. Repair protein persistence at DNA lesions characterizes XPF defect with Cockayne syndrome features. Nucleic Acids Res 2018; 46:9563-9577. [PMID: 30165384 PMCID: PMC6182131 DOI: 10.1093/nar/gky774] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/19/2018] [Accepted: 08/16/2018] [Indexed: 12/28/2022] Open
Abstract
The structure-specific ERCC1-XPF endonuclease plays a key role in DNA damage excision by nucleotide excision repair (NER) and interstrand crosslink repair. Mutations in this complex can either cause xeroderma pigmentosum (XP) or XP combined with Cockayne syndrome (XPCS-complex) or Fanconi anemia. However, most patients carry compound heterozygous mutations, which confounds the dissection of the phenotypic consequences for each of the identified XPF alleles. Here, we analyzed the functional impact of individual pathogenic XPF alleles on NER. We show that XP-causing mutations diminish XPF recruitment to DNA damage and only mildly affect global genome NER. In contrast, an XPCS-complex-specific mutation causes persistent recruitment of XPF and the upstream core NER machinery to DNA damage and severely impairs both global genome and transcription-coupled NER. Remarkably, persistence of NER factors at DNA damage appears to be a common feature of XPCS-complex cells, suggesting that this could be a determining factor contributing to the development of additional developmental and/or neurodegenerative features in XP patients.
Collapse
Affiliation(s)
- Mariangela Sabatella
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Arjan F Theil
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Cristina Ribeiro-Silva
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Jana Slyskova
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Karen Thijssen
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Chantal Voskamp
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Hannes Lans
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| | - Wim Vermeulen
- Department of Molecular Genetics, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, University Erasmus Medical Center Rotterdam, 3000 CA, The Netherlands
| |
Collapse
|
23
|
Sharp SP, Malizia RA, Walrath T, D'Souza SS, Booth CJ, Kartchner BJ, Lee EC, Stain SC, O'Connor W. DNA damage response genes mark the early transition from colitis to neoplasia in colitis-associated colon cancer. Gene 2018; 677:299-307. [PMID: 30121380 DOI: 10.1016/j.gene.2018.08.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/04/2018] [Indexed: 12/12/2022]
Abstract
Chronic intestinal inflammation predisposes patients with Inflammatory Bowel Disease (IBD) to Colitis-Associated Cancer (CAC). In the setting of chronic inflammation, microsatellite instability (MSI) results from early loss of DNA damage response (DDR) genes, ultimately leading to tumor formation. Despite continued efforts to improve early detection of high risk, pre-dysplastic regions in IBD patients, current macroscopic and genetic surveillance modalities remain limited. Therefore, understanding the regulation of key DDR genes in the progression from colitis to cancer may improve molecular surveillance of CAC. To evaluate DDR gene regulation in the transition from colitis to tumorigenesis, we utilized the well-established Azoxymethane/Dextran Sodium Sulfate (AOM/DSS) pre-clinical murine model of CAC in C57BL/6 mice. In order to assess colonic tumor burden in the setting of mutagen and intestinal irritation, tumors were visualized and graded in real time through high-resolution murine colonoscopy. Upon sacrifice, colons were opened and assessed for macroscopic tumor via high magnification surgical lenses (HMSL). Tissues were then sectioned and separated into groups based on the presence or absence of macroscopically visible tumor. Critical DDR genes were evaluated by semi-quantitative RT-PCR. Interestingly, colon tissue with macroscopically visible tumor (MVT) and colon tissue prior to observable tumor (the non-macroscopically visible tumor-developing group, NMVT) were identical in reduced mRNA expression of mlh1, anapc1, and ercc4 relative to colitic mice without mutagen, or those receiving mutagen alone. Colitis alone was sufficient to reduce colonic ercc4 expression when compared to NMVT mice. Therefore, reduced ercc4 expression may mark the early transition to CAC in a pre-clinical model, with expression reduced prior to the onset of observable tumor. Moreover, the expression of select DDR genes inversely correlated with chronicity of inflammatory disease. These data suggest ercc4 expression may define early stages in the progression to CAC.
Collapse
Affiliation(s)
- Stephen P Sharp
- Department of Surgery, Albany Medical College, Albany, NY, USA.
| | | | - Travis Walrath
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| | - Shanti S D'Souza
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| | - Carmen J Booth
- Department of Comparative Medicine, Yale University, New Haven, CT, USA.
| | - Brittany J Kartchner
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| | - Edward C Lee
- Department of Surgery, Albany Medical College, Albany, NY, USA.
| | - Steven C Stain
- Department of Surgery, Albany Medical College, Albany, NY, USA.
| | - William O'Connor
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
24
|
Zhuo ZJ, Liu W, Zhang J, Zhu J, Zhang R, Tang J, Yang T, Zou Y, He J, Xia H. Functional Polymorphisms at ERCC1/XPF Genes Confer Neuroblastoma Risk in Chinese Children. EBioMedicine 2018; 30:113-119. [PMID: 29544698 PMCID: PMC5952228 DOI: 10.1016/j.ebiom.2018.03.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/05/2018] [Accepted: 03/05/2018] [Indexed: 02/07/2023] Open
Abstract
Variations in nucleotide excision repair pathway genes may predispose to initiation of cancers. However, polymorphisms of ERCC1/XPF genes and neuroblastoma risk have not been investigated before. To evaluate the relevance of polymorphisms of ERCC1/XPF genes in influencing neuroblastoma susceptibility, we genotyped four polymorphisms in ERCC1/XPF genes using a Chinese population of 393 cases and 812 controls. The results showed that ERCC1 rs2298881 and rs11615 predisposed to enhanced neuroblastoma risk [CA vs. AA: adjusted odds ratio (OR)=1.94, 95% confidence interval (CI)=1.30-2.89, P=0.0012; CC vs. AA: adjusted OR=2.18, 95% CI=1.45-3.26, P=0.0002 for rs2298881, and AG vs. GG: adjusted OR=1.31, 95% CI=1.02-1.69, P=0.038 for rs11615]. Moreover, XPF rs2276466 was also associated with increased neuroblastoma risk (GG vs. CC: adjusted OR=1.66, 95% CI=1.02-2.71, P=0.043). In the combined analysis of ERCC1, we found that carriers with 2-3 risk genotypes were more likely to get risk of neuroblastoma, when compared to those with 0-1 risk genotype (adjusted OR=1.75; 95% CI=1.25-2.45, P=0.0012). Our study indicates that common genetic variations in ERCC1/XPF genes predispose to neuroblastoma risk, which needs to be further validated by ongoing efforts.
Collapse
Affiliation(s)
- Zhen-Jian Zhuo
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China; School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wei Liu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jiao Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jinhong Zhu
- Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Ruizhong Zhang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jue Tang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Tianyou Yang
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yan Zou
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China.
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China.
| |
Collapse
|
25
|
Chinen LTD, Abdallah EA, Braun AC, Flores BDCTDCP, Corassa M, Sanches SM, Fanelli MF. Circulating Tumor Cells as Cancer Biomarkers in the Clinic. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:1-41. [PMID: 28560666 DOI: 10.1007/978-3-319-55947-6_1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is believed that the development of metastatic cancer requires the presence of circulating tumor cells (CTCs) , which are found in a patient's circulation as rare abnormal cells comingled with billions of the normal red and white blood cells. The systems developed for detection of CTCs have brought progress to cancer treatment. The molecular characterization of CTCs can aid in the development of new drugs, and their presence during treatment can help clinicians determine the prognosis of the patient. Studies have been carried out in patients early in the disease course, with only primary tumors, and the role of CTCs in prognosis seems to be as important as it is in patients with metastatic disease. The published studies on CTCs have focused on their prognostic significance, their utility in real-time monitoring of therapies, the identification of therapeutic and resistance targets, and understanding the process of metastasis . The analysis of CTCs during the early stages, as a "liquid biopsy," helps to monitor patients at different points in the disease course, including minimal residual disease, providing valuable information about the very early assessment of treatment effectiveness. Finally, CTCs can be used to screen patients with family histories of cancer or with diseases that can lead to the development of cancer. With standard protocols, this easily obtained and practical tool can be used to prevent the growth and spread of cancer. In this chapter, we review some important aspects of CTCs , surveying the disease aspects where these cells have been investigated.
Collapse
Affiliation(s)
| | - Emne Ali Abdallah
- International Research Center, A. C. Camargo Cancer Center, Rua Taguá 440, São Paulo, SP, 01508-010, Brazil
| | - Alexcia Camila Braun
- International Research Center, A. C. Camargo Cancer Center, Rua Taguá 440, São Paulo, SP, 01508-010, Brazil
| | | | - Marcelo Corassa
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| | - Solange Moraes Sanches
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| | - Marcello Ferretti Fanelli
- Department of Medical Oncology, A. C. Camargo Cancer Center, Rua Professor Antônio Prudente, São Paulo, SP, 01509-010, Brazil
| |
Collapse
|
26
|
Brosey CA, Ahmed Z, Lees-Miller SP, Tainer JA. What Combined Measurements From Structures and Imaging Tell Us About DNA Damage Responses. Methods Enzymol 2017; 592:417-455. [PMID: 28668129 DOI: 10.1016/bs.mie.2017.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
DNA damage outcomes depend upon the efficiency and fidelity of DNA damage responses (DDRs) for different cells and damage. As such, DDRs represent tightly regulated prototypical systems for linking nanoscale biomolecular structure and assembly to the biology of genomic regulation and cell signaling. However, the dynamic and multifunctional nature of DDR assemblies can render elusive the correlation between the structures of DDR factors and specific biological disruptions to the DDR when these structures are altered. In this chapter, we discuss concepts and strategies for combining structural, biophysical, and imaging techniques to investigate DDR recognition and regulation, and thus bridge sequence-level structural biochemistry to quantitative biological outcomes visualized in cells. We focus on representative DDR responses from PARP/PARG/AIF damage signaling in DNA single-strand break repair and nonhomologous end joining complexes in double-strand break repair. Methods with exemplary experimental results are considered with a focus on strategies for probing flexibility, conformational changes, and assembly processes that shape a predictive understanding of DDR mechanisms in a cellular context. Integration of structural and imaging measurements promises to provide foundational knowledge to rationally control and optimize DNA damage outcomes for synthetic lethality and for immune activation with resulting insights for biology and cancer interventions.
Collapse
Affiliation(s)
- Chris A Brosey
- The University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| | - Zamal Ahmed
- The University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| | - Susan P Lees-Miller
- Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
| | - John A Tainer
- The University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
27
|
Xiao M, Xiao S, Straaten TVD, Xue P, Zhang G, Zheng X, Zhang Q, Cai Y, Jin C, Yang J, Wu S, Zhu G, Lu X. Genetic polymorphisms in 19q13.3 genes associated with alteration of repair capacity to BPDE-DNA adducts in primary cultured lymphocytes. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 812:39-47. [PMID: 27908386 DOI: 10.1016/j.mrgentox.2016.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 10/18/2016] [Accepted: 10/25/2016] [Indexed: 12/27/2022]
Abstract
Benzo[a]pyrene(B[a]P), and its ultimate metabolite Benzo[a]pyrene 7,8-diol 9,10-epoxide (BPDE), are classic DNA damaging carcinogens. DNA damage in cells caused by BPDE is normally repaired by Nucleotide Excision Repair (NER) and Base Excision Repair (BER). Genetic variations in NER and BER can change individual DNA repair capacity to DNA damage induced by BPDE. In the present study we determined the number of in vitro induced BPDE-DNA adducts in lymphocytes, to reflect individual susceptibility to Polycyclic aromatic hydrocarbons (PAHs)-induced carcinogenesis. The BPDE-DNA adduct level in lymphocytes were assessed by high performance liquid chromatography (HPLC) in 281 randomly selected participants. We genotyped for 9 single nucleotide polymorphisms (SNPs) in genes involved in NER (XPB rs4150441, XPC rs2228001, rs2279017 and XPF rs4781560), BER (XRCC1 rs25487, rs25489 and rs1799782) and genes located on chromosome 19q13.2-3 (PPP1R13L rs1005165 and CAST rs967591). We found that 3 polymorphisms in chromosome 19q13.2-3 were associated with lower levels of BPDE-DNA adducts (MinorT allele in XRCC1 rs1799782, minor T allele in PPP1R13L rs1005165 and minor A allele in CAST rs967571). In addition, a modified comet assay was performed to further confirm the above conclusions. We found both minor T allele in PPP1R13L rs1005165 and minor A allele in CAST rs967571 were associated with the lower levels of BPDE-adducts. Our data suggested that the variant genotypes of genes in chromosome 19q13.2-3 are associated with the alteration of repair efficiency to DNA damage caused by Benzo[a]pyrene, and may contribute to enhance predictive value for individual's DNA repair capacity in response to environmental carcinogens.
Collapse
Affiliation(s)
- Mingyang Xiao
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Sha Xiao
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Tahar van der Straaten
- Dept. Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ping Xue
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Guopei Zhang
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Xiao Zheng
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Qianye Zhang
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Yuan Cai
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Cuihong Jin
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Jinghua Yang
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Shengwen Wu
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Guolian Zhu
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China
| | - Xiaobo Lu
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, PR China.
| |
Collapse
|
28
|
Wolf Horrell EM, Boulanger MC, D’Orazio JA. Melanocortin 1 Receptor: Structure, Function, and Regulation. Front Genet 2016; 7:95. [PMID: 27303435 PMCID: PMC4885833 DOI: 10.3389/fgene.2016.00095] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/13/2016] [Indexed: 01/04/2023] Open
Abstract
The melanocortin 1 receptor (MC1R) is a melanocytic Gs protein coupled receptor that regulates skin pigmentation, UV responses, and melanoma risk. It is a highly polymorphic gene, and loss of function correlates with a fair, UV-sensitive, and melanoma-prone phenotype due to defective epidermal melanization and sub-optimal DNA repair. MC1R signaling, achieved through adenylyl cyclase activation and generation of the second messenger cAMP, is hormonally controlled by the positive agonist melanocortin, the negative agonist agouti signaling protein, and the neutral antagonist β-defensin 3. Activation of cAMP signaling up-regulates melanin production and deposition in the epidermis which functions to limit UV penetration into the skin and enhances nucleotide excision repair (NER), the genomic stability pathway responsible for clearing UV photolesions from DNA to avoid mutagenesis. Herein we review MC1R structure and function and summarize our laboratory's findings on the molecular mechanisms by which MC1R signaling impacts NER.
Collapse
Affiliation(s)
- Erin M. Wolf Horrell
- Department of Physiology, University of Kentucky College of MedicineLexington, KY, USA
| | - Mary C. Boulanger
- Markey Cancer Center, University of Kentucky College of MedicineLexington, KY, USA
| | - John A. D’Orazio
- Department of Physiology, University of Kentucky College of MedicineLexington, KY, USA
- Markey Cancer Center, University of Kentucky College of MedicineLexington, KY, USA
- Departments of Pediatrics, Toxicology and Cancer Biology, Physiology, and Pharmacology and Nutritional Sciences, University of Kentucky College of MedicineLexington, KY, USA
| |
Collapse
|
29
|
Su CH, Cheng C, Tzeng TY, Lin IH, Hsu MT. An H2A Histone Isotype, H2ac, Associates with Telomere and Maintains Telomere Integrity. PLoS One 2016; 11:e0156378. [PMID: 27228173 PMCID: PMC4882029 DOI: 10.1371/journal.pone.0156378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/13/2016] [Indexed: 11/19/2022] Open
Abstract
Telomeres are capped at the ends of eukaryotic chromosomes and are composed of TTAGGG repeats bound to the shelterin complex. Here we report that a replication-dependent histone H2A isotype, H2ac, was associated with telomeres in human cells and co-immunoprecipitates with telomere repeat factor 2 (TRF2) and protection of telomeres protein 1 (POT1), whereas other histone H2A isotypes and mutations of H2ac did not bind to telomeres or these two proteins. The amino terminal basic domain of TRF2 was necessary for the association with H2ac and for the recruitment of H2ac to telomeres. Depletion of H2ac led to loss of telomeric repeat sequences, the appearance of dysfunctional telomeres, and chromosomal instability, including chromosomal breaks and anaphase bridges, as well as accumulation of telomere-associated DNA damage factors in H2ac depleted cells. Additionally, knockdown of H2ac elicits an ATM-dependent DNA damage response at telomeres and depletion of XPF protects telomeres against H2ac-deficiency-induced G-strand overhangs loss and DNA damage response, and prevents chromosomal instability. These findings suggest that the H2A isotype, H2ac, plays an essential role in maintaining telomere functional integrity.
Collapse
Affiliation(s)
- Chia-Hsin Su
- Institute of Biochemistry and Molecular Biology, School of Life Science, National Yang-Ming University, Taipei, 11221, Taiwan, Republic of China
| | - Ching Cheng
- Institute of Biochemistry and Molecular Biology, School of Life Science, National Yang-Ming University, Taipei, 11221, Taiwan, Republic of China
| | - Tsai-Yu Tzeng
- VYM Genome Research Center, National Yang-Ming University, University System of Taiwan, Taipei, 11221, Taiwan, Republic of China
| | - I-Hsuan Lin
- Institute of Biochemistry and Molecular Biology, School of Life Science, National Yang-Ming University, Taipei, 11221, Taiwan, Republic of China
| | - Ming-Ta Hsu
- Institute of Biochemistry and Molecular Biology, School of Life Science, National Yang-Ming University, Taipei, 11221, Taiwan, Republic of China
- VYM Genome Research Center, National Yang-Ming University, University System of Taiwan, Taipei, 11221, Taiwan, Republic of China
- Chien-Tien Hsu Cancer Research Foundation, Taipei, 11221, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
30
|
Bautista-Niño PK, Portilla-Fernandez E, Vaughan DE, Danser AHJ, Roks AJM. DNA Damage: A Main Determinant of Vascular Aging. Int J Mol Sci 2016; 17:E748. [PMID: 27213333 PMCID: PMC4881569 DOI: 10.3390/ijms17050748] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 01/16/2023] Open
Abstract
Vascular aging plays a central role in health problems and mortality in older people. Apart from the impact of several classical cardiovascular risk factors on the vasculature, chronological aging remains the single most important determinant of cardiovascular problems. The causative mechanisms by which chronological aging mediates its impact, independently from classical risk factors, remain to be elucidated. In recent years evidence has accumulated that unrepaired DNA damage may play an important role. Observations in animal models and in humans indicate that under conditions during which DNA damage accumulates in an accelerated rate, functional decline of the vasculature takes place in a similar but more rapid or more exaggerated way than occurs in the absence of such conditions. Also epidemiological studies suggest a relationship between DNA maintenance and age-related cardiovascular disease. Accordingly, mouse models of defective DNA repair are means to study the mechanisms involved in biological aging of the vasculature. We here review the evidence of the role of DNA damage in vascular aging, and present mechanisms by which genomic instability interferes with regulation of the vascular tone. In addition, we present potential remedies against vascular aging induced by genomic instability. Central to this review is the role of diverse types of DNA damage (telomeric, non-telomeric and mitochondrial), of cellular changes (apoptosis, senescence, autophagy), mediators of senescence and cell growth (plasminogen activator inhibitor-1 (PAI-1), cyclin-dependent kinase inhibitors, senescence-associated secretory phenotype (SASP)/senescence-messaging secretome (SMS), insulin and insulin-like growth factor 1 (IGF-1) signaling), the adenosine monophosphate-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR)-nuclear factor kappa B (NFκB) axis, reactive oxygen species (ROS) vs. endothelial nitric oxide synthase (eNOS)-cyclic guanosine monophosphate (cGMP) signaling, phosphodiesterase (PDE) 1 and 5, transcription factor NF-E2-related factor-2 (Nrf2), and diet restriction.
Collapse
Affiliation(s)
- Paula K Bautista-Niño
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Eliana Portilla-Fernandez
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Douglas E Vaughan
- Department of Medicine & Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - A H Jan Danser
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands.
| |
Collapse
|
31
|
Li F, Xie X, Ren X, Zhang J. A meta-analysis identifies ERCC1 gene polymorphism as a predictor of better patient response to treatment with radiochemotherapy. Cancer Chemother Pharmacol 2016; 77:1183-91. [PMID: 27100737 DOI: 10.1007/s00280-016-3015-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/16/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE This meta-analysis aimed to evaluate whether an association exists between the ERCC1 rs11615 (C>T) polymorphism and patient response to platinum-based chemotherapy and radiation-based chemotherapy. METHODS Publications were selected from PubMed and MEDLINE. A meta-analysis was conducted to determine the association between genetic polymorphisms and response to platinum-based chemotherapy and radiation-based chemotherapy by checking the odds ratios (ORs) and 95 % confidence intervals (CIs). RESULTS In our overall analysis, the ERCC1 rs11615 (C>T) polymorphism was not associated with response to platinum-based chemotherapy in five comparison models. In the subgroup analyses by ethnicity, the ERCC1 rs11615 (C>T) polymorphism was shown to be significantly associated with objective response in Caucasian patients treated with platinum-based chemotherapy in the recessive model (TT vs. CT/CC OR 0.696, 95 % CI 0.508-0.954, heterogeneity = 0.330), but the association was not observed in the Asian population. The C allele was significantly associated with better response to radiochemotherapy in the recessive model comparison (TT vs. CC/CT OR 0.724, 95 % CI 0.585-0.869, heterogeneity = 0.008). Subgroup analysis by cancer type revealed that the C allele of ERCC1 rs11615 predicted a better response in esophageal cancers in two comparison models (T vs. C: OR 0.756, 95 % CI 0.648-0.880, heterogeneity = 0.653; TT vs. TC/CC OR 0.457, 95 % CI 0.306-0.684, heterogeneity = 0.723). Stratified analysis by ethnicity showed a better response in Caucasians in allelic comparison model (T vs. C: OR 0.895, 95 % CI 0.819-0.977, heterogeneity = 0.095). CONCLUSION Together, our results suggest that the ERCC1 rs11615 (C>T) polymorphism was associated with therapeutic response in Caucasian patients and C allele of ERCC1 rs11615 could represent a genetic molecular marker to predict better patient response to radiochemotherapy in recessive model. However, larger prospective randomized trials will be required.
Collapse
Affiliation(s)
- Fengying Li
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qing-chun East Road, Hangzhou, 310016, People's Republic of China
| | - Xinyou Xie
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qing-chun East Road, Hangzhou, 310016, People's Republic of China
| | - Xiaobin Ren
- Department of Prevention and Control of Infectious Diseases, Hangzhou Municipal Center for Disease Control and Prevention, 568 Mingshi Road, Hangzhou, 310021, People's Republic of China.
| | - Jun Zhang
- Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qing-chun East Road, Hangzhou, 310016, People's Republic of China.
| |
Collapse
|
32
|
Ma Y, Deng XD, Feng Y, Zhang W, Wang SX, Liu Y, Liu H. Association of XPF Levels and Genetic Polymorphism with Susceptibility to Ischemic Stroke. J Mol Neurosci 2016; 59:168-76. [DOI: 10.1007/s12031-016-0729-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/02/2016] [Indexed: 11/28/2022]
|
33
|
Abstract
Nucleotide excision repair (NER) is a highly versatile and efficient DNA repair process, which is responsible for the removal of a large number of structurally diverse DNA lesions. Its extreme broad substrate specificity ranges from DNA damages formed upon exposure to ultraviolet radiation to numerous bulky DNA adducts induced by mutagenic environmental chemicals and cytotoxic drugs used in chemotherapy. Defective NER leads to serious diseases, such as xeroderma pigmentosum (XP). Eight XP complementation groups are known of which seven (XPA-XPG) are caused by mutations in genes involved in the NER process. The eighth gene, XPV, codes for the DNA polymerase ɳ, which replicates through DNA lesions in a process called translesion synthesis (TLS). Over the past decade, detailed structural information of these DNA repair proteins involved in eukaryotic NER and TLS have emerged. These structures allow us now to understand the molecular mechanism of the NER and TLS processes in quite some detail and we have begun to understand the broad substrate specificity of NER. In this review, we aim to highlight recent advances in the process of damage recognition and repair as well as damage tolerance by the XP proteins.
Collapse
|
34
|
Zessin PJM, Sporbert A, Heilemann M. PCNA appears in two populations of slow and fast diffusion with a constant ratio throughout S-phase in replicating mammalian cells. Sci Rep 2016; 6:18779. [PMID: 26758689 PMCID: PMC4725372 DOI: 10.1038/srep18779] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 11/26/2015] [Indexed: 01/14/2023] Open
Abstract
DNA replication is a fundamental cellular process that precedes cell division. Proliferating cell nuclear antigen (PCNA) is a central scaffold protein that orchestrates DNA replication by recruiting many factors essential for the replication machinery. We studied the mobility of PCNA in live mammalian cells using single-particle tracking in combination with photoactivated-localization microscopy (sptPALM) and found two populations. The first population which is only present in cells with active DNA replication, showed slow diffusion and was found to be located in replication foci. The second population showed fast diffusion, and represents the nucleoplasmic pool of unbound PCNA not involved in DNA replication. The ratio of these two populations remained constant throughout different stages of S-phase. A fraction of molecules in both populations showed spatially constrained mobility. We determined an exploration radius of ~100 nm for 13% of the slow-diffusing PCNA molecules, and of ~600 nm for 46% of the fast-diffusing PCNA molecules.
Collapse
Affiliation(s)
- Patrick J M Zessin
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Anje Sporbert
- Advanced Light Microscopy, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt/Main, Germany
| |
Collapse
|
35
|
Li XY, Huang J, Jiang HX, Du YC, Han GM, Kong DM. Molecular logic gates based on DNA tweezers responsive to multiplex restriction endonucleases. RSC Adv 2016. [DOI: 10.1039/c6ra05132d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Self-assembled DNA tweezers containing four different restriction endonuclease recognition sites were designed and a set of logic gates were constructed.
Collapse
Affiliation(s)
- Xiao-Yu Li
- State Key Laboratory of Medicinal Chemical Biology
- College of Chemistry
- Nankai University Tianjin
- People's Republic of China
| | - Juan Huang
- State Key Laboratory of Medicinal Chemical Biology
- College of Chemistry
- Nankai University Tianjin
- People's Republic of China
| | - Hong-Xin Jiang
- State Key Laboratory of Medicinal Chemical Biology
- College of Chemistry
- Nankai University Tianjin
- People's Republic of China
| | - Yi-Chen Du
- State Key Laboratory of Medicinal Chemical Biology
- College of Chemistry
- Nankai University Tianjin
- People's Republic of China
| | - Gui-Mei Han
- State Key Laboratory of Medicinal Chemical Biology
- College of Chemistry
- Nankai University Tianjin
- People's Republic of China
| | - De-Ming Kong
- State Key Laboratory of Medicinal Chemical Biology
- College of Chemistry
- Nankai University Tianjin
- People's Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| |
Collapse
|
36
|
Fadda E. Role of the XPA protein in the NER pathway: A perspective on the function of structural disorder in macromolecular assembly. Comput Struct Biotechnol J 2015; 14:78-85. [PMID: 26865925 PMCID: PMC4710682 DOI: 10.1016/j.csbj.2015.11.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/23/2022] Open
Abstract
Lack of structure is often an essential functional feature of protein domains. The coordination of macromolecular assemblies in DNA repair pathways is yet another task disordered protein regions are highly implicated in. Here I review the available experimental and computational data and within this context discuss the functional role of structure and disorder in one of the essential scaffolding proteins in the nucleotide excision repair (NER) pathway, namely Xeroderma pigmentosum complementation group A (XPA). From the analysis of the current knowledge, in addition to protein–protein docking and secondary structure prediction results presented for the first time herein, a mechanistic framework emerges, where XPA builds the NER pre-incision complex in a modular fashion, as “beads on a string”, where the protein–protein interaction “beads”, or modules, are interconnected by disordered link regions. This architecture is ideal to avoid the expected steric hindrance constraints of the DNA expanded bubble. Finally, the role of the XPA structural disorder in binding affinity modulation and in the sequential binding of NER core factors in the pre-incision complex is also discussed.
Collapse
Affiliation(s)
- Elisa Fadda
- Department of Chemistry, Maynooth University, Maynooth, Kildare, Ireland
| |
Collapse
|
37
|
SUMO and ubiquitin-dependent XPC exchange drives nucleotide excision repair. Nat Commun 2015; 6:7499. [PMID: 26151477 PMCID: PMC4501428 DOI: 10.1038/ncomms8499] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023] Open
Abstract
XPC recognizes UV-induced DNA lesions and initiates their removal by nucleotide excision repair (NER). Damage recognition in NER is tightly controlled by ubiquitin and SUMO modifications. Recent studies have shown that the SUMO-targeted ubiquitin ligase RNF111 promotes K63-linked ubiquitylation of SUMOylated XPC after DNA damage. However, the exact regulatory function of these modifications in vivo remains elusive. Here we show that RNF111 is required for efficient repair of ultraviolet-induced DNA lesions. RNF111-mediated ubiquitylation promotes the release of XPC from damaged DNA after NER initiation, and is needed for stable incorporation of the NER endonucleases XPG and ERCC1/XPF. Our data suggest that RNF111, together with the CRL4(DDB2) ubiquitin ligase complex, is responsible for sequential XPC ubiquitylation, which regulates the recruitment and release of XPC and is crucial for efficient progression of the NER reaction, thereby providing an extra layer of quality control of NER.
Collapse
|
38
|
Abstract
The nucleus is physically distinct from the cytoplasm in ways that suggest new ideas and approaches for interrogating the operation of this organelle. Chemical bond formation and breakage underlie the lives of cells, but as this special issue of Molecular Biology of the Cell attests, the nonchemical aspects of cell nuclei present a new frontier to biologists and biophysicists.
Collapse
Affiliation(s)
- Thoru Pederson
- Program in Cell and Developmental Dynamics, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605
| | - John F Marko
- Department of Molecular Biosciences and Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208
| |
Collapse
|
39
|
Mendenhall AR, Tedesco PM, Sands B, Johnson TE, Brent R. Single Cell Quantification of Reporter Gene Expression in Live Adult Caenorhabditis elegans Reveals Reproducible Cell-Specific Expression Patterns and Underlying Biological Variation. PLoS One 2015; 10:e0124289. [PMID: 25946008 PMCID: PMC4422670 DOI: 10.1371/journal.pone.0124289] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/07/2015] [Indexed: 11/22/2022] Open
Abstract
In multicellular organisms such as Caenorhabditis elegans, differences in complex phenotypes such as lifespan correlate with the level of expression of particular engineered reporter genes. In single celled organisms, quantitative understanding of responses to extracellular signals and of cell-to-cell variation in responses has depended on precise measurement of reporter gene expression. Here, we developed microscope-based methods to quantify reporter gene expression in cells of Caenorhabditis elegans with low measurement error. We then quantified expression in strains that carried different configurations of Phsp-16.2-fluorescent-protein reporters, in whole animals, and in all 20 cells of the intestine tissue, which is responsible for most of the fluorescent signal. Some animals bore more recently developed single copy Phsp-16.2 reporters integrated at defined chromosomal sites, others, “classical” multicopy reporter gene arrays integrated at random sites. At the level of whole animals, variation in gene expression was similar: strains with single copy reporters showed the same amount of animal-to-animal variation as strains with multicopy reporters. At the level of cells, in animals with single copy reporters, the pattern of expression in cells within the tissue was highly stereotyped. In animals with multicopy reporters, the cell-specific expression pattern was also stereotyped, but distinct, and somewhat more variable. Our methods are rapid and gentle enough to allow quantification of expression in the same cells of an animal at different times during adult life. They should allow investigators to use changes in reporter expression in single cells in tissues as quantitative phenotypes, and link those to molecular differences. Moreover, by diminishing measurement error, they should make possible dissection of the causes of the remaining, real, variation in expression. Understanding such variation should help reveal its contribution to differences in complex phenotypic outcomes in multicellular organisms.
Collapse
Affiliation(s)
- Alexander R. Mendenhall
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail: (RB); (ARM)
| | - Patricia M. Tedesco
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado, United States of America
| | - Bryan Sands
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Thomas E. Johnson
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado, United States of America
- Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States of America
- Biofrontiers Institute, University of Colorado, Boulder, Colorado, United States of America
| | - Roger Brent
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail: (RB); (ARM)
| |
Collapse
|
40
|
Geverts B, van Royen ME, Houtsmuller AB. Analysis of biomolecular dynamics by FRAP and computer simulation. Methods Mol Biol 2015; 1251:109-33. [PMID: 25391797 DOI: 10.1007/978-1-4939-2080-8_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We present a Monte Carlo simulation environment for modelling complex biological molecular interaction networks and for the design, validation, and quantitative analysis of FRAP assays to study these. The program is straightforward in its implementation and can be instructed through an intuitive script language. The simulation tool fits very well in a systems biology research setting that aims to maintain an interactive cycle of experiment-driven modelling and model-driven experimentation: the model and the experiment are in the same simulation. The full program can be obtained by request to the authors.
Collapse
Affiliation(s)
- Bart Geverts
- Department of Pathology, Josephine Nefkens Institute, Erasmus Optical Imaging Centre, Erasmus MC, Postbus 2040, Rotterdam, 3000 CA, The Netherlands
| | | | | |
Collapse
|
41
|
Qiu J, Wang X, Meng X, Zheng Y, Li G, Ma J, Ye G, Li Y, Li J. Attenuated NER expressions of XPF and XPC associated with smoking are involved in the recurrence of bladder cancer. PLoS One 2014; 9:e115224. [PMID: 25535740 PMCID: PMC4275238 DOI: 10.1371/journal.pone.0115224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 11/20/2014] [Indexed: 01/01/2023] Open
Abstract
The varied NER genes and smoking are two important risk factors of bladder cancer, but the mechanism of the NER protein and smoking in cancer progression, however, remains unclear. In this report, we compared the expressions of NER genes in 79 bladder cancer tissues with or without any recurrence by real-time PCR and then analyzed the varied NER genes by immunochemistry in 219 bladder cancer tissue samples. Based on the clinical data, we analyzed the clinical value of varied NER genes and smoking in 219 bladder cancers by the Kaplan-Meier method and Cox proportional hazards regression. We found the expressions of the NER gene XPF and XPC were significantly lower in bladder cancer tissues with a recurrence compared with those without a recurrence at mRNA level. Also, the patients with the XPF and XPC defect had a statistically significant lower median recurrence-free survival time than those without the XPF and XPC defect, and smoking can make this difference more remarkable. Our results suggest that XPF and XPC expression may be a potential predictive factor for bladder cancer, and smoking can not only influence the recurrence of bladder cancer as a single factor but also aggravate the results of the XPF defect and XPC defect.
Collapse
Affiliation(s)
- Jianhong Qiu
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Xiangwei Wang
- Department of Urology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaodong Meng
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Yan Zheng
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Gang Li
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Jiyao Ma
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Gang Ye
- Department of Urology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yong Li
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Jie Li
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
- * E-mail:
| |
Collapse
|
42
|
Song C, Zhang Q, Han GM, Du YC, Kong DM. A facile fluorescence method for endonuclease detection using exonuclease III-aided signal amplification of a molecular beacon. RSC Adv 2014. [DOI: 10.1039/c4ra09676b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
43
|
Rapsomaniki MA, Cinquemani E, Giakoumakis NN, Kotsantis P, Lygeros J, Lygerou Z. Inference of protein kinetics by stochastic modeling and simulation of fluorescence recovery after photobleaching experiments. ACTA ACUST UNITED AC 2014; 31:355-62. [PMID: 25273108 DOI: 10.1093/bioinformatics/btu619] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
MOTIVATION Fluorescence recovery after photobleaching (FRAP) is a functional live cell imaging technique that permits the exploration of protein dynamics in living cells. To extract kinetic parameters from FRAP data, a number of analytical models have been developed. Simplifications are inherent in these models, which may lead to inexhaustive or inaccurate exploitation of the experimental data. An appealing alternative is offered by the simulation of biological processes in realistic environments at a particle level. However, inference of kinetic parameters using simulation-based models is still limited. RESULTS We introduce and demonstrate a new method for the inference of kinetic parameter values from FRAP data. A small number of in silico FRAP experiments is used to construct a mapping from FRAP recovery curves to the parameters of the underlying protein kinetics. Parameter estimates from experimental data can then be computed by applying the mapping to the observed recovery curves. A bootstrap process is used to investigate identifiability of the physical parameters and determine confidence regions for their estimates. Our method circumvents the computational burden of seeking the best-fitting parameters via iterative simulation. After validation on synthetic data, the method is applied to the analysis of the nuclear proteins Cdt1, PCNA and GFPnls. Parameter estimation results from several experimental samples are in accordance with previous findings, but also allow us to discuss identifiability issues as well as cell-to-cell variability of the protein kinetics. IMPLEMENTATION All methods were implemented in MATLAB R2011b. Monte Carlo simulations were run on the HPC cluster Brutus of ETH Zurich. CONTACT lygeros@control.ee.ethz.ch or lygerou@med.upatras.gr SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Maria Anna Rapsomaniki
- Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France
| | - Eugenio Cinquemani
- Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France
| | - Nickolaos Nikiforos Giakoumakis
- Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France
| | - Panagiotis Kotsantis
- Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France
| | - John Lygeros
- Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France
| | - Zoi Lygerou
- Department of Biology, School of Medicine, University of Patras, 26505, Rio, Patras, Greece, Institut für Automatik, ETH Zürich, 8092 Zürich, Switzerland and INRIA Grenoble-Rhône-Alpes, Montbonnot, 38334 Saint-Ismier Cedex, France
| |
Collapse
|
44
|
Bendtsen KM, Jensen MB, May A, Rasmussen LJ, Trusina A, Bohr VA, Jensen MH. Dynamics of the DNA repair proteins WRN and BLM in the nucleoplasm and nucleoli. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2014; 43:509-16. [PMID: 25119658 DOI: 10.1007/s00249-014-0981-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/20/2014] [Accepted: 07/21/2014] [Indexed: 11/30/2022]
Abstract
We have investigated the mobility of two EGFP-tagged DNA repair proteins, WRN and BLM. In particular, we focused on the dynamics in two locations, the nucleoli and the nucleoplasm. We found that both WRN and BLM use a "DNA-scanning" mechanism, with rapid binding-unbinding to DNA resulting in effective diffusion. In the nucleoplasm WRN and BLM have effective diffusion coefficients of 1.62 and 1.34 μm(2)/s, respectively. Likewise, the dynamics in the nucleoli are also best described by effective diffusion, but with diffusion coefficients a factor of ten lower than in the nucleoplasm. From this large reduction in diffusion coefficient we were able to classify WRN and BLM as DNA damage scanners. In addition to WRN and BLM we also classified other DNA damage proteins and found they all fall into one of two categories. Either they are scanners, similar to WRN and BLM, with very low diffusion coefficients, suggesting a scanning mechanism, or they are almost freely diffusing, suggesting that they interact with DNA only after initiation of a DNA damage response.
Collapse
|
45
|
Xu W, Deng R, Wang L, Li J. Multiresponsive Rolling Circle Amplification for DNA Logic Gates Mediated by Endonuclease. Anal Chem 2014; 86:7813-8. [DOI: 10.1021/ac501726s] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Weidong Xu
- Department
of Chemistry,
Beijing Key Laboratory for Analytical Methods and Instrumentation, Tsinghua University, Beijing 100084, China
| | - Ruijie Deng
- Department
of Chemistry,
Beijing Key Laboratory for Analytical Methods and Instrumentation, Tsinghua University, Beijing 100084, China
| | - Lida Wang
- Department
of Chemistry,
Beijing Key Laboratory for Analytical Methods and Instrumentation, Tsinghua University, Beijing 100084, China
| | - Jinghong Li
- Department
of Chemistry,
Beijing Key Laboratory for Analytical Methods and Instrumentation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
46
|
Kuhlmann JD, Wimberger P, Bankfalvi A, Keller T, Schöler S, Aktas B, Buderath P, Hauch S, Otterbach F, Kimmig R, Kasimir-Bauer S. ERCC1-positive circulating tumor cells in the blood of ovarian cancer patients as a predictive biomarker for platinum resistance. Clin Chem 2014; 60:1282-9. [PMID: 25015375 DOI: 10.1373/clinchem.2014.224808] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Platinum resistance constitutes one of the most recognized clinical challenges for ovarian cancer. Notably, the detection of the primary tumor-based excision repair cross-complementation group 1 (ERCC1) protein by immunohistochemistry was recently shown to be inaccurate for the prediction of platinum resistance. On the basis of the previous finding that circulating tumor cells (CTC) in the blood of ovarian cancer patients are prognostically significant, and given our hypothesis that the negative prognostic impact of CTC may arise from a cellular phenotype associated with platinum resistance, we asked whether expression of the excision repair cross-complementation group 1 (ERCC1) gene in the form of the ERCC1 transcript in CTC may be a suitable blood-based biomarker for platinum resistance. METHODS The presence of CTC was analyzed by immunomagnetic CTC enrichment (n = 143 patients) targeting the epithelial epitopes epithelial cell adhesion molecule (EPCAM) (also known as GA733-2) and mucin 1, cell surface associated (MUC1), followed by multiplex reverse-transcription PCR to detect the transcripts EPCAM, MUC1, and mucin 16, cell surface associated (MUC16) (also known as CA125), including ERCC1 transcripts in a separate approach. ERCC1 expression in primary tumors was comparatively assessed by immunohistochemistry, using the antibody 8F1. RESULTS At primary diagnosis, the presence of CTC was observed in 14% of patients and constituted an independent predictor of overall survival (OS) (P = 0.041). ERCC1-positive CTC (ERCC1(+)CTC) were observed in 8% of patients and constituted an independent predictor, not only for OS but also for progression-free survival (PFS) (P = 0.026 and P = 0.009, respectively). More interestingly, we discovered the presence of ERCC1(+)CTC at primary diagnosis to be likewise an independent predictor of platinum resistance (P = 0.010), whereas ERCC1 expression in corresponding primary tumor tissue predicted neither platinum resistance nor prognosis. CONCLUSIONS The presence of ERCC1(+)CTC can serve as a blood-based diagnostic biomarker for predicting platinum resistance at primary diagnosis of ovarian cancer.
Collapse
Affiliation(s)
- Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, West German Cancer Center, University Hospital of Essen, Essen, Germany; Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany;
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, West German Cancer Center, University Hospital of Essen, Essen, Germany; Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Agnes Bankfalvi
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Pathology and Neuropathology, University Hospital of Essen, Essen, Germany
| | | | - Sarah Schöler
- Department of Pathology and Neuropathology, University Hospital of Essen, Essen, Germany
| | - Bahriye Aktas
- Department of Gynecology and Obstetrics, West German Cancer Center, University Hospital of Essen, Essen, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul Buderath
- Department of Gynecology and Obstetrics, West German Cancer Center, University Hospital of Essen, Essen, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Friedrich Otterbach
- Department of Pathology and Neuropathology, University Hospital of Essen, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, West German Cancer Center, University Hospital of Essen, Essen, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, West German Cancer Center, University Hospital of Essen, Essen, Germany; German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
47
|
Liccardi G, Hartley JA, Hochhauser D. Importance of EGFR/ERCC1 interaction following radiation-induced DNA damage. Clin Cancer Res 2014; 20:3496-506. [PMID: 24780295 DOI: 10.1158/1078-0432.ccr-13-2695] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The epidermal growth factor receptor (EGFR) plays an important role in cellular response to chemotherapy and radiotherapy through modulation of DNA repair. EGFR activates DNA-dependent protein kinase (DNA-PK) stimulating repair of DNA strand breaks (SB) and interstrand crosslinks (ICL). We investigated the role of EGFR in repair of ionizing radiation (IR)-induced SB independently of DNA-PK. EXPERIMENTAL DESIGN The EGFR interactome was investigated via mass spectrometry. IR-induced EGFR-ERCC1 binding was validated biochemically and via proximity ligation assay in different cell lines including the M059K and M059J glioma cell lines, proficient and deficient for the expression of DNAPKcs, respectively. EGFR-ERCC1 functional significance following IR-induced SB was investigated in knockdown experiments with the Comet and γH2AX foci assays. The effect of this interaction was tested with EGFR-ERCC1 knockdown in combination with gefitinib and NU7026 using the MTT and apoptosis assays. RESULTS This study demonstrates that EGFR inhibition further impairs IR-induced DNA repair in cells lacking expression of DNAPKcs or in combination with the DNAPK inhibitor NU7026. Our data suggest a role for EGFR in DNA repair independent of DNAPKcs but dependent on ERCC1. Alkaline comet and γH2AX foci assays in cells depleted of EGFR, ERCC1, or EGFR-ERCC1 expression demonstrated involvement of this interaction in DNA repair. Cellular survival and apoptosis data correlate with levels of residual DNA damage underlying the importance of this complex following SB. CONCLUSION These data emphasize the importance of understanding the various mechanisms by which EGFR modulates DNA repair to optimize targeted therapy for patients with cancer.
Collapse
Affiliation(s)
- Gianmaria Liccardi
- Authors' Affiliation: Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, University College London, London, United Kingdom
| | - John A Hartley
- Authors' Affiliation: Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, University College London, London, United Kingdom
| | - Daniel Hochhauser
- Authors' Affiliation: Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
48
|
Raj DDA, Jaarsma D, Holtman IR, Olah M, Ferreira FM, Schaafsma W, Brouwer N, Meijer MM, de Waard MC, van der Pluijm I, Brandt R, Kreft KL, Laman JD, de Haan G, Biber KPH, Hoeijmakers JHJ, Eggen BJL, Boddeke HWGM. Priming of microglia in a DNA-repair deficient model of accelerated aging. Neurobiol Aging 2014; 35:2147-60. [PMID: 24799273 DOI: 10.1016/j.neurobiolaging.2014.03.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/21/2014] [Accepted: 03/23/2014] [Indexed: 12/16/2022]
Abstract
Aging is associated with reduced function, degenerative changes, and increased neuroinflammation of the central nervous system (CNS). Increasing evidence suggests that changes in microglia cells contribute to the age-related deterioration of the CNS. The most prominent age-related change of microglia is enhanced sensitivity to inflammatory stimuli, referred to as priming. It is unclear if priming is due to intrinsic microglia ageing or induced by the ageing neural environment. We have studied this in Ercc1 mutant mice, a DNA repair-deficient mouse model that displays features of accelerated aging in multiple tissues including the CNS. In Ercc1 mutant mice, microglia showed hallmark features of priming such as an exaggerated response to peripheral lipopolysaccharide exposure in terms of cytokine expression and phagocytosis. Specific targeting of the Ercc1 deletion to forebrain neurons resulted in a progressive priming response in microglia exemplified by phenotypic alterations. Summarizing, these data show that neuronal genotoxic stress is sufficient to switch microglia from a resting to a primed state.
Collapse
Affiliation(s)
- Divya D A Raj
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Dick Jaarsma
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Inge R Holtman
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marta Olah
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Filipa M Ferreira
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wandert Schaafsma
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michel M Meijer
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Monique C de Waard
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ingrid van der Pluijm
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Renata Brandt
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Karim L Kreft
- Department of Immunology, Erasmus University Medical Center and MS Center ErasMS, Rotterdam, the Netherlands; Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jon D Laman
- Department of Immunology, Erasmus University Medical Center and MS Center ErasMS, Rotterdam, the Netherlands
| | - Gerald de Haan
- Department of Cell Biology, European Research Institute on the Biology of Aging, University of Groningen, University Medical Center, Groningen, the Netherlands
| | - Knut P H Biber
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Psychiatry and Psychotherapy, University Medical Center, Freiburg, Germany
| | - Jan H J Hoeijmakers
- Department of Genetics, Cancer Genomics Center, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hendrikus W G M Boddeke
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
49
|
Uphoff S, Kapanidis AN. Studying the organization of DNA repair by single-cell and single-molecule imaging. DNA Repair (Amst) 2014; 20:32-40. [PMID: 24629485 PMCID: PMC4119245 DOI: 10.1016/j.dnarep.2014.02.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 02/09/2014] [Accepted: 02/14/2014] [Indexed: 12/26/2022]
Abstract
Single-cell experiments to study stochastic events and heterogeneity in DNA repair. Quantifying DNA repair protein concentration, diffusion, and localization in cells. Direct observation of DNA repair using photoactivated single-molecule tracking.
DNA repair safeguards the genome against a diversity of DNA damaging agents. Although the mechanisms of many repair proteins have been examined separately in vitro, far less is known about the coordinated function of the whole repair machinery in vivo. Furthermore, single-cell studies indicate that DNA damage responses generate substantial variation in repair activities across cells. This review focuses on fluorescence imaging methods that offer a quantitative description of DNA repair in single cells by measuring protein concentrations, diffusion characteristics, localizations, interactions, and enzymatic rates. Emerging single-molecule and super-resolution microscopy methods now permit direct visualization of individual proteins and DNA repair events in vivo. We expect much can be learned about the organization of DNA repair by linking cell heterogeneity to mechanistic observations at the molecular level.
Collapse
Affiliation(s)
- Stephan Uphoff
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom.
| | - Achillefs N Kapanidis
- Biological Physics Research Group, Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, United Kingdom.
| |
Collapse
|
50
|
Van Royen ME, van Cappellen WA, Geverts B, Schmidt T, Houtsmuller AB, Schaaf MJM. Androgen receptor complexes probe DNA for recognition sequences by short random interactions. J Cell Sci 2014; 127:1406-16. [PMID: 24481814 DOI: 10.1242/jcs.135228] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Owing to the tremendous progress in microscopic imaging of fluorescently labeled proteins in living cells, the insight into the highly dynamic behavior of transcription factors has rapidly increased over the past decade. However, a consistent quantitative scheme of their action is still lacking. Using the androgen receptor (AR) as a model system, we combined three different fluorescence microscopy assays: single-molecule microscopy, photobleaching and correlation spectroscopy, to provide a quantitative model of the action of this transcription factor. This approach enabled us to distinguish two types of AR-DNA binding: very brief interactions, in the order of a few hundred milliseconds, and hormone-induced longer-lasting interactions, with a characteristic binding time of several seconds. In addition, freely mobile ARs were slowed down in the presence of hormone, suggesting the formation of large AR-co-regulator complexes in the nucleoplasm upon hormone activation. Our data suggest a model in which mobile hormone-induced complexes of transcription factors and co-regulators probe DNA by briefly binding at random sites, only forming relatively stable transcription initiation complexes when bound to specific recognition sequences.
Collapse
|