Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 56(1): 78-92.e6, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36630919

ABSTRACT

Tissue repair processes maintain proper organ function following mechanical or infection-related damage. In addition to antibacterial properties, mucosal associated invariant T (MAIT) cells express a tissue repair transcriptomic program and promote skin wound healing when expanded. Herein, we use a human-like mouse model of full-thickness skin excision to assess the underlying mechanisms of MAIT cell tissue repair function. Single-cell RNA sequencing analysis suggested that skin MAIT cells already express a repair program at steady state. Following skin excision, MAIT cells promoted keratinocyte proliferation, thereby accelerating healing. Using skin grafts, parabiosis, and adoptive transfer experiments, we show that MAIT cells migrated into the wound in a T cell receptor (TCR)-independent but CXCR6 chemokine receptor-dependent manner. Amphiregulin secreted by MAIT cells following excision promoted wound healing. Expression of the repair function was probably independent of sustained TCR stimulation. Overall, our study provides mechanistic insights into MAIT cell wound healing function in the skin.


Subject(s)
Amphiregulin , Histocompatibility Antigens Class I , Mucosal-Associated Invariant T Cells , Wound Healing , Animals , Humans , Mice , Amphiregulin/metabolism , Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens , Mucosal-Associated Invariant T Cells/metabolism , Receptors, Antigen, T-Cell/metabolism
2.
Mol Cell ; 82(4): 728-740, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34965379

ABSTRACT

Ferroptosis is a unique type of non-apoptotic cell death resulting from the unrestrained occurrence of peroxidized phospholipids, which are subject to iron-mediated production of lethal oxygen radicals. This cell death modality has been detected across many organisms, including in mammals, where it can be used as a defense mechanism against pathogens or even harnessed by T cells to sensitize tumor cells toward effective killing. Conversely, ferroptosis is considered one of the main cell death mechanisms promoting degenerative diseases. Emerging evidence suggests that ferroptosis represents a vulnerability in certain cancers. Here, we critically review recent advances linking ferroptosis vulnerabilities of dedifferentiating and persister cancer cells to the dependency of these cells on iron, a potential Achilles heel for small-molecule intervention. We provide a perspective on the mechanisms reliant on iron that contribute to the persister cancer cell state and how this dependency may be exploited for therapeutic benefits.


Subject(s)
Ferroptosis , Iron/metabolism , Lipid Peroxidation , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Differentiation , Ferroptosis/drug effects , Homeostasis , Humans , Lipid Peroxidation/drug effects , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Signal Transduction
3.
Nature ; 617(7960): 386-394, 2023 May.
Article in English | MEDLINE | ID: mdl-37100912

ABSTRACT

Inflammation is a complex physiological process triggered in response to harmful stimuli1. It involves cells of the immune system capable of clearing sources of injury and damaged tissues. Excessive inflammation can occur as a result of infection and is a hallmark of several diseases2-4. The molecular bases underlying inflammatory responses are not fully understood. Here we show that the cell surface glycoprotein CD44, which marks the acquisition of distinct cell phenotypes in the context of development, immunity and cancer progression, mediates the uptake of metals including copper. We identify a pool of chemically reactive copper(II) in mitochondria of inflammatory macrophages that catalyses NAD(H) redox cycling by activating hydrogen peroxide. Maintenance of NAD+ enables metabolic and epigenetic programming towards the inflammatory state. Targeting mitochondrial copper(II) with supformin (LCC-12), a rationally designed dimer of metformin, induces a reduction of the NAD(H) pool, leading to metabolic and epigenetic states that oppose macrophage activation. LCC-12 interferes with cell plasticity in other settings and reduces inflammation in mouse models of bacterial and viral infections. Our work highlights the central role of copper as a regulator of cell plasticity and unveils a therapeutic strategy based on metabolic reprogramming and the control of epigenetic cell states.


Subject(s)
Cell Plasticity , Copper , Inflammation , Signal Transduction , Animals , Mice , Copper/metabolism , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , NAD/metabolism , Signal Transduction/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Hydrogen Peroxide/metabolism , Epigenesis, Genetic/drug effects , Metformin/analogs & derivatives , Oxidation-Reduction , Cell Plasticity/drug effects , Cell Plasticity/genetics , Macrophage Activation/drug effects , Macrophage Activation/genetics
4.
Chembiochem ; 25(9): e202400211, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38530090

ABSTRACT

This symposium is the 5th PSL (Paris Sciences & Lettres) Chemical Biology meeting (2015, 2016, 2019, 2023, 2024) held at Institut Curie. This initiative originally started at Institut de Chimie des Substances Naturelles (ICSN) in Gif-sur-Yvette, with a strong focus on chemistry. It was then continued at the Institut Curie (2015) covering a larger scope, before becoming the official PSL Chemical Biology meeting. This latest edition hosted around 150 participants and was focused on the burgeoning field of ferroptosis, its mechanism and implications in health and disease. While not initially planned, it was felt that the next large Ferroptosis venue (CSHA, China) would not happen before late 2024. A discussion involving Conrad, Birsoy, Ubellacker, Brabletz and Rodriguez next to lake Como in Italy sponsored by the DKFZ, prompted us to fill in this gap and to organize a Ferroptosis meeting in Paris beforehand.


Subject(s)
Ferroptosis , Animals , Humans , Ferroptosis/drug effects
5.
Chembiochem ; 24(8): e202300093, 2023 04 17.
Article in English | MEDLINE | ID: mdl-36942862

ABSTRACT

This symposium is the third PSL (Paris Sciences & Lettres) Chemical Biology meeting (2016, 2019, 2023) held at Institut Curie. This initiative originally started at Institut de Chimie des Substances Naturelles (ICSN) in Gif-sur-Yvette (2013, 2014), under the directorship of Professor Max Malacria, with a strong focus on chemistry. It was then continued at the Institut Curie (2015) covering a larger scope, before becoming the official PSL Chemical Biology meeting. This latest edition was postponed twice for the reasons that we know. This has given us the opportunity to invite additional speakers of great standing. This year, Institut Curie hosted around 300 participants, including 220 on site and over 80 online. The pandemic has had, at least, the virtue of promoting online meetings, which we came to realize is not perfect but has its own merits. In particular, it enables those with restricted time and resources to take part in events and meetings, which can now accommodate unlimited participants. We apologize to all those who could not attend in person this time due to space limitation at Institut Curie.


Subject(s)
Biology , Humans , Paris
6.
J Am Chem Soc ; 144(26): 11536-11545, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35696539

ABSTRACT

Persister cancer cells represent rare populations of cells resistant to therapy. Cancer cells can exploit epithelial-mesenchymal plasticity to adopt a drug-tolerant state that does not depend on genetic alterations. Small molecules that can interfere with cell plasticity or kill cells in a cell state-dependent manner are highly sought after. Salinomycin has been shown to kill cancer cells in the mesenchymal state by sequestering iron in lysosomes, taking advantage of the iron addiction of this cell state. Here, we report the chemo- and stereoselective synthesis of a series of structurally complex small molecule chimeras of salinomycin derivatives and the iron-reactive dihydroartemisinin. We show that these chimeras accumulate in lysosomes and can react with iron to release bioactive species, thereby inducing ferroptosis in drug-tolerant pancreatic cancer cells and biopsy-derived organoids of pancreatic ductal adenocarcinoma. This work paves the way toward the development of new cancer medicines acting through active ferroptosis.


Subject(s)
Ferroptosis , Pancreatic Neoplasms , Prodrugs , Humans , Iron , Pancreatic Neoplasms/drug therapy , Prodrugs/pharmacology , Reactive Oxygen Species , Pancreatic Neoplasms
7.
Angew Chem Int Ed Engl ; 61(32): e202205231, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35612562

ABSTRACT

Interferons (IFN) are cytokines which, upon binding to cell surface receptors, trigger a series of downstream biochemical events including Janus Kinase (JAK) activation, phosphorylation of Signal Transducer and Activator of Transcription protein (STAT), translocation of pSTAT to the nucleus and transcriptional activation. Dysregulated IFN signalling has been linked to cancer progression and auto-immune diseases. Here, we report the serendipitous discovery of a small molecule that blocks IFNγ activation of JAK-STAT signalling. Further lead optimisation gave rise to a potent and more selective analogue that exerts its activity by a mechanism consistent with direct IFNγ targeting in vitro, which reduces bleeding in model of haemorrhagic colitis in vivo. This first-in-class small molecule also inhibits type I and III IFN-induced STAT phosphorylation in vitro. Our work provides the basis for the development of pan-IFN inhibitory drugs.


Subject(s)
Interferons , Janus Kinases , Interferon-gamma , Interferons/metabolism , Interferons/pharmacology , Phosphorylation , Signal Transduction
8.
Nat Chem Biol ; 15(5): 549, 2019 05.
Article in English | MEDLINE | ID: mdl-30833779

ABSTRACT

In the version of this article originally published, several co-authors had incorrect affiliation footnote numbers listed in the author list. Tatiana Cañeque and Angelica Mariani should each have affiliation numbers 3, 4 and 5, and Emmanuelle Charafe-Jauffret should have number 6. Additionally, there was an extra space in the name of co-author Robert P. St.Onge. These errors have been corrected in the HTML and PDF versions of the paper and the Supplementary Information PDF.

9.
Nat Chem Biol ; 15(4): 358-366, 2019 04.
Article in English | MEDLINE | ID: mdl-30742123

ABSTRACT

Peripheral membrane proteins orchestrate many physiological and pathological processes, making regulation of their activities by small molecules highly desirable. However, they are often refractory to classical competitive inhibition. Here, we demonstrate that potent and selective inhibition of peripheral membrane proteins can be achieved by small molecules that target protein-membrane interactions by a noncompetitive mechanism. We show that the small molecule Bragsin inhibits BRAG2-mediated Arf GTPase activation in vitro in a manner that requires a membrane. In cells, Bragsin affects the trans-Golgi network in a BRAG2- and Arf-dependent manner. The crystal structure of the BRAG2-Bragsin complex and structure-activity relationship analysis reveal that Bragsin binds at the interface between the PH domain of BRAG2 and the lipid bilayer to render BRAG2 unable to activate lipidated Arf. Finally, Bragsin affects tumorsphere formation in breast cancer cell lines. Bragsin thus pioneers a novel class of drugs that function by altering protein-membrane interactions without disruption.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/physiology , ADP-Ribosylation Factor 1/metabolism , Cell Line, Tumor , GTP Phosphohydrolases , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , HeLa Cells , Humans , Lipid Bilayers , Membrane Glycoproteins/metabolism , Nucleotides , Pleckstrin Homology Domains/physiology , Protein Binding , Signal Transduction , Structure-Activity Relationship , Sulfotransferases/metabolism
10.
Adv Exp Med Biol ; 1301: 81-121, 2021.
Article in English | MEDLINE | ID: mdl-34370289

ABSTRACT

Ferroptosis is a dedicated mode of cell death involving iron, reactive oxygen species and lipid peroxidation. Involved in processes such as glutathione metabolism, lysosomal iron retention or interference with lipid metabolism, leading either to activation or inhibition of ferroptosis. Given the implications of ferroptosis in diseases such as cancer, aging, Alzheimer and infectious diseases, new molecular mechanisms underlying ferroptosis and small molecules regulators that target those mechanisms have prompted a great deal of interest. Here, we discuss the current scenario of small molecules modulating ferroptosis and critically assess what is known about their mechanisms of action.


Subject(s)
Ferroptosis , Cell Death , Humans , Iron , Lipid Peroxidation , Reactive Oxygen Species
11.
Chemistry ; 26(33): 7369-7373, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32083771

ABSTRACT

Cancer stem cells (CSC) constitute a cell subpopulation in solid tumors that is responsible for resistance to conventional chemotherapy, metastasis and cancer relapse. The natural product Salinomycin can selectively target this cell niche by directly interacting with lysosomal iron, taking advantage of upregulated iron homeostasis in CSC. Here, inhibitors of the divalent metal transporter 1 (DMT1) have been identified that selectively target CSC by blocking lysosomal iron translocation. This leads to lysosomal iron accumulation, production of reactive oxygen species and cell death with features of ferroptosis. DMT1 inhibitors selectively target CSC in primary cancer cells and circulating tumor cells, demonstrating the physiological relevance of this strategy. Taken together, this opens up opportunities to tackle unmet needs in anti-cancer therapy.


Subject(s)
Cation Transport Proteins/chemistry , Iron/chemistry , Lysosomes/chemistry , Neoplastic Stem Cells/chemistry , Pyrans/chemistry , Reactive Oxygen Species/chemistry , Cation Transport Proteins/metabolism , Cell Death , Homeostasis , Humans , Iron/metabolism , Lysosomes/metabolism , Neoplastic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation
12.
Chemistry ; 26(33): 7416-7424, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32083773

ABSTRACT

Salinomycin (1) exhibits a large spectrum of biological activities including the capacity to selectively eradicate cancer stem cells (CSC), making it and its derivatives promising candidates for the development of drug leads against CSC. It has been previously shown that salinomycin and its C20-propargylamine derivative (Ironomycin (2)) accumulate in lysosomes and sequester iron in this organelle. Herein, a library of salinomycin derivatives is reported, including products of C20-amination, C1-esterification, C9-oxidation, and C28-dehydration. The biological activity of these compounds is evaluated against transformed human mammary epithelial HMLER CD24low /CD44high cells, a well-established model of breast CSC, and HMLER CD24high /CD44low cells deprived of CSC properties. Unlike other structural alterations, derivative 4, which displays a cyclopropylamine at position C20, showed a strikingly low IC50 value of 23 nm against HMLER CD24low /CD44high cells. This study provides highly selective molecules to target the CSC niche, a potential interesting advance for drug development to prevent cancer resistance.


Subject(s)
Breast Neoplasms/drug therapy , Hyaluronan Receptors/chemistry , Iron/agonists , Lysosomes/chemistry , Neoplastic Stem Cells/chemistry , Pyrans/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Hyaluronan Receptors/metabolism , Lysosomes/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pyrans/chemistry
13.
Nat Rev Genet ; 15(12): 783-96, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25311424

ABSTRACT

Small molecules--including various approved and novel cancer therapeutics--can operate at the genomic level by targeting the DNA and protein components of chromatin. Emerging evidence suggests that functional interactions between small molecules and the genome are non-stochastic and are influenced by a dynamic interplay between DNA sequences and chromatin states. The establishment of genome-wide maps of small-molecule targets using unbiased methodologies can help to characterize and exploit drug responses. In this Review, we discuss how high-throughput sequencing strategies, such as ChIP-seq (chromatin immunoprecipitation followed by sequencing) and Chem-seq (chemical affinity capture and massively parallel DNA sequencing), are enabling the comprehensive identification of small-molecule target sites throughout the genome, thereby providing insights into unanticipated drug effects.


Subject(s)
Drug Delivery Systems , Gene Expression Regulation, Neoplastic/genetics , Animals , Chromatin Immunoprecipitation , Epigenesis, Genetic/drug effects , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, DNA , Small Molecule Libraries
14.
Angew Chem Int Ed Engl ; 59(14): 5721-5729, 2020 03 27.
Article in English | MEDLINE | ID: mdl-31769920

ABSTRACT

Chemical proteomics is widely applied in small-molecule target identification. However, in general it does not identify non-protein small-molecule targets, and thus, alternative methods for target identification are in high demand. We report the discovery of the autophagy inhibitor autoquin and the identification of its molecular mode of action using image-based morphological profiling in the cell painting assay. A compound-induced fingerprint representing changes in 579 cellular parameters revealed that autoquin accumulates in lysosomes and inhibits their fusion with autophagosomes. In addition, autoquin sequesters Fe2+ in lysosomes, resulting in an increase of lysosomal reactive oxygen species and ultimately cell death. Such a mechanism of action would have been challenging to unravel by current methods. This work demonstrates the potential of the cell painting assay to deconvolute modes of action of small molecules, warranting wider application in chemical biology.


Subject(s)
Autophagy , Iron/metabolism , Lysosomes/metabolism , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cinchona Alkaloids/chemistry , Cinchona Alkaloids/pharmacology , Humans , Microscopy, Fluorescence , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
15.
Chembiochem ; 20(7): 968-973, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30803119

ABSTRACT

Chemical Biology is the science of designing chemical tools to dissect and manipulate biology at different scales. It provides the fertile ground from which to address important problems of our society, such as human health and environment.


Subject(s)
Biology , Chemistry , Humans , Paris
16.
Biochemistry ; 56(38): 5125-5133, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28858527

ABSTRACT

Arf GTPases and their guanine nucleotide exchange factors (ArfGEFs) are major regulators of membrane traffic and organelle structure in cells. They are associated with a variety of diseases and are thus attractive therapeutic targets for inhibition by small molecules. Several inhibitors of unrelated chemical structures have been discovered, which have shown their potential in dissecting molecular pathways and blocking disease-related functions. However, their specificity across the ArfGEF family has remained elusive. Importantly, inhibitory responses in the context of membranes, which are critical determinants of Arf and ArfGEF cellular functions, have not been investigated. Here, we compare the efficiency and specificity of four structurally distinct ArfGEF inhibitors, Brefeldin A, SecinH3, M-COPA, and NAV-2729, toward six ArfGEFs (human ARNO, EFA6, BIG1, and BRAG2 and Legionella and Rickettsia RalF). Inhibition was assessed by fluorescence kinetics using pure proteins, and its modulation by membranes was determined with lipidated GTPases in the presence of liposomes. Our analysis shows that despite the intra-ArfGEF family resemblance, each inhibitor has a specific inhibitory profile. Notably, M-COPA is a potent pan-ArfGEF inhibitor, and NAV-2729 inhibits all GEFs, the strongest effects being against BRAG2 and Arf1. Furthermore, the presence of the membrane-binding domain in Legionella RalF reveals a strong inhibitory effect of BFA that is not measured on its GEF domain alone. This study demonstrates the value of family-wide assays with incorporation of membranes, and it should enable accurate dissection of Arf pathways by these inhibitors to best guide their use and development as therapeutic agents.


Subject(s)
Brefeldin A/pharmacology , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Naphthols/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidinones/pharmacology , Triazoles/pharmacology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/antagonists & inhibitors , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Cell Membrane , Chlorobenzenes , Fluorescence , GTPase-Activating Proteins/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Liposomes/chemistry , Solutions
17.
J Am Chem Soc ; 139(4): 1400-1403, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28094937

ABSTRACT

Enoxacin is a small molecule that stimulates RNA interference (RNAi) and acts as a growth inhibitor selectively in cancer but not in untransformed cells. Here, we used alkenox, a clickable enoxacin surrogate, coupled with quantitative mass spectrometry, to identify PIWIL3 as a mechanistic target of enoxacin. PIWIL3 is an Argonaute protein of the PIWI subfamily that is mainly expressed in the germline and that mediates RNAi through piRNAs. Our results suggest that cancer cells re-express PIWIL3 to repress RNAi through miRNAs and thus open a new opportunity for cancer-specific targeting.


Subject(s)
Argonaute Proteins/analysis , Breast Neoplasms/drug therapy , Enoxacin/pharmacology , Argonaute Proteins/antagonists & inhibitors , Argonaute Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Enoxacin/chemistry , Female , Humans , MCF-7 Cells , Mass Spectrometry , Molecular Structure
18.
Chembiochem ; 18(10): 883-887, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28371105

ABSTRACT

Chemical biology, the science of understanding biological processes at the molecular level, has grown exponentially with the development of chemical strategies to manipulate and quantify biology with unprecedented precision. Recent advances presented at the Université Paris Sciences et Lettres symposium are discussed.


Subject(s)
Biology , Chemistry , Congresses as Topic , Drug Discovery/methods , Humans , Molecular Probes
19.
Bioorg Med Chem Lett ; 27(2): 203-207, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27919658

ABSTRACT

We have synthesized a collection of quinolizinium fluorescent dyes for the purpose of cell imaging. Preliminary biological studies in human U2OS osteosarcoma cancer cells have shown that different functional groups appended to the cationic quinolizinium scaffold efficiently modulate photophysical properties but also cellular distribution. While quinolizinium probes are known nuclear staining reagents, we have identified a particular quinolizinium derivative salt that targets the lysosomal compartment. This finding raises the question of predictability of specific organelle targeting from structural features of small molecules.


Subject(s)
Fluorescent Dyes/pharmacology , Quinolizines/pharmacology , Anthraquinones/metabolism , Artemisinins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Endocytosis/drug effects , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Hydrogen-Ion Concentration , Intercalating Agents/chemical synthesis , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Lysosomes/metabolism , Molecular Imaging , Quinolizines/chemical synthesis , Quinolizines/chemistry
20.
Angew Chem Int Ed Engl ; 56(23): 6483-6487, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28474855

ABSTRACT

Cisplatin derivatives can form various types of DNA lesions (DNA-Pt) and trigger pleiotropic DNA damage responses. Here, we report a strategy to visualize DNA-Pt with high resolution, taking advantage of a novel azide-containing derivative of cisplatin we named APPA, a cellular pre-extraction protocol and the labeling of DNA-Pt by means of click chemistry in cells. Our investigation revealed that pretreating cells with the histone deacetylase (HDAC) inhibitor SAHA led to detectable clusters of DNA-Pt that colocalized with the ubiquitin ligase RAD18 and the replication protein PCNA. Consistent with activation of translesion synthesis (TLS) under these conditions, SAHA and cisplatin cotreatment promoted focal accumulation of the low-fidelity polymerase Polη that also colocalized with PCNA. Remarkably, these cotreatments synergistically triggered mono-ubiquitination of PCNA and apoptosis in a RAD18-dependent manner. Our data provide evidence for a role of chromatin in regulating genome targeting with cisplatin derivatives and associated cellular responses.


Subject(s)
Antineoplastic Agents/pharmacology , Chromatin/physiology , Cisplatin/pharmacology , Genome, Human/drug effects , Cell Line, Tumor , Cisplatin/analogs & derivatives , Click Chemistry , DNA/drug effects , DNA Damage , DNA-Directed DNA Polymerase/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Molecular Probes , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL