146 results on '"Oscar Fernandez-Capetillo"'
Search Results
2. New regulators of the tetracycline‐inducible gene expression system identified by chemical and genetic screens
- Author
-
Valeria Colicchia, Maria Häggblad, Oleksandra Sirozh, Bartlomiej Porebski, Mirela Balan, Xuexin Li, Louise Lidemalm, Jordi Carreras‐Puigvert, Daniela Hühn, and Oscar Fernandez‐Capetillo
- Subjects
ALS ,chemical screen ,doxycycline ,TDP‐43 ,tetR ,Biology (General) ,QH301-705.5 - Abstract
The tetracycline repressor (tetR)‐regulated system is a widely used tool to specifically control gene expression in mammalian cells. Based on this system, we generated a human osteosarcoma cell line, which allows for the inducible expression of an EGFP fusion of the TAR DNA‐binding protein 43 (TDP‐43), which has been linked to neurodegenerative diseases. Consistent with previous findings, TDP‐43 overexpression led to the accumulation of aggregates and limited the viability of U2OS. Using this inducible system, we conducted a chemical screen with a library that included FDA‐approved drugs. While the primary screen identified several compounds that prevented TDP‐43 toxicity, further experiments revealed that these chemicals abrogated the doxycycline‐dependent TDP‐43 expression. This antagonistic effect was observed with both doxycycline and tetracycline, and in several Tet‐On cell lines expressing different genes, confirming the general effect of these compounds as inhibitors of the tetR system. Using the same cell line, a genome‐wide CRISPR/Cas9 screen identified epigenetic regulators such as the G9a methyltransferase and TRIM28 as potential modifiers of TDP‐43 toxicity. Yet again, further experiments revealed that G9a inhibition or TRIM28 loss prevented doxycycline‐dependent expression of TDP‐43. In summary, we have identified new chemical and genetic regulators of the tetR system, thereby raising awareness of the limitations of this approach to conduct chemical or genetic screening in mammalian cells.
- Published
- 2022
- Full Text
- View/download PDF
3. Activation of the integrated stress response is a vulnerability for multidrug‐resistant FBXW7‐deficient cells
- Author
-
Laura Sanchez‐Burgos, Belén Navarro‐González, Santiago García‐Martín, Oleksandra Sirozh, Jorge Mota‐Pino, Elena Fueyo‐Marcos, Héctor Tejero, Marta Elena Antón, Matilde Murga, Fátima Al‐Shahrour, and Oscar Fernandez‐Capetillo
- Subjects
drug resistance ,FBXW7 ,GCN2 ,ISR ,mitochondria ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract FBXW7 is one of the most frequently mutated tumor suppressors, deficiency of which has been associated with resistance to some anticancer therapies. Through bioinformatics and genome‐wide CRISPR screens, we here reveal that FBXW7 deficiency leads to multidrug resistance (MDR). Proteomic analyses found an upregulation of mitochondrial factors as a hallmark of FBXW7 deficiency, which has been previously linked to chemotherapy resistance. Despite this increased expression of mitochondrial factors, functional analyses revealed that mitochondria are under stress, and genetic or chemical targeting of mitochondria is preferentially toxic for FBXW7‐deficient cells. Mechanistically, the toxicity of therapies targeting mitochondrial translation such as the antibiotic tigecycline relates to the activation of the integrated stress response (ISR) in a GCN2 kinase‐dependent manner. Furthermore, the discovery of additional drugs that are toxic for FBXW7‐deficient cells showed that all of them unexpectedly activate a GCN2‐dependent ISR regardless of their accepted mechanism of action. Our study reveals that while one of the most frequent mutations in cancer reduces the sensitivity to the vast majority of available therapies, it renders cells vulnerable to ISR‐activating drugs.
- Published
- 2022
- Full Text
- View/download PDF
4. A bispecific monomeric nanobody induces spike trimer dimers and neutralizes SARS-CoV-2 in vivo
- Author
-
Leo Hanke, Hrishikesh Das, Daniel J. Sheward, Laura Perez Vidakovics, Egon Urgard, Ainhoa Moliner-Morro, Changil Kim, Vivien Karl, Alec Pankow, Natalie L. Smith, Bartlomiej Porebski, Oscar Fernandez-Capetillo, Erdinc Sezgin, Gabriel K. Pedersen, Jonathan M. Coquet, B. Martin Hällberg, Ben Murrell, and Gerald M. McInerney
- Subjects
Science - Abstract
Here, the authors isolate and characterize a bispecific monomeric nanobody that induces dimerization of SARS-CoV-2 spike trimers, neutralizes variants of concerns as well as SARS-CoV, and inhibits SARS-CoV-2 infection in mice.
- Published
- 2022
- Full Text
- View/download PDF
5. Prolonged estrogen deprivation triggers a broad immunosuppressive phenotype in breast cancer cells
- Author
-
Daniela Hühn, Pablo Martí‐Rodrigo, Silvana Mouron, Catherine Hansel, Kirsten Tschapalda, Bartlomiej Porebski, Maria Häggblad, Louise Lidemalm, Miguel Quintela‐Fandino, Jordi Carreras‐Puigvert, and Oscar Fernandez‐Capetillo
- Subjects
breast cancer ,estrogen receptor ,HLA ,immunotherapy ,inflammation ,PD‐L1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Among others, expression levels of programmed cell death 1 ligand 1 (PD‐L1) have been explored as biomarkers of the response to immune checkpoint inhibitors in cancer therapy. Here, we present the results of a chemical screen that interrogated how medically approved drugs influence PD‐L1 expression. As expected, corticosteroids and inhibitors of Janus kinases were among the top PD‐L1 downregulators. In addition, we identified that PD‐L1 expression is induced by antiestrogenic compounds. Transcriptomic analyses indicate that chronic estrogen receptor alpha (ERα) inhibition triggers a broad immunosuppressive program in ER‐positive breast cancer cells, which is subsequent to their growth arrest and involves the activation of multiple immune checkpoints together with the silencing of the antigen‐presenting machinery. Accordingly, estrogen‐deprived MCF7 cells are resistant to T‐cell‐mediated cell killing, in a manner that is independent of PD‐L1, but which is reverted by estradiol. Our study reveals that while antiestrogen therapies efficiently limit the growth of ER‐positive breast cancer cells, they concomitantly trigger a transcriptional program that favors their immune evasion.
- Published
- 2022
- Full Text
- View/download PDF
6. An in silico analysis identifies drugs potentially modulating the cytokine storm triggered by SARS-CoV-2 infection
- Author
-
Laura Sanchez-Burgos, Gonzalo Gómez-López, Fátima Al-Shahrour, and Oscar Fernandez-Capetillo
- Subjects
Medicine ,Science - Abstract
Abstract The ongoing COVID-19 pandemic is one of the biggest health challenges of recent decades. Among the causes of mortality triggered by SARS-CoV-2 infection, the development of an inflammatory “cytokine storm” (CS) plays a determinant role. Here, we used transcriptomic data from the bronchoalveolar lavage fluid (BALF) of COVID-19 patients undergoing a CS to obtain gene-signatures associated to this pathology. Using these signatures, we interrogated the Connectivity Map (CMap) dataset that contains the effects of over 5000 small molecules on the transcriptome of human cell lines, and looked for molecules which effects on transcription mimic or oppose those of the CS. As expected, molecules that potentiate immune responses such as PKC activators are predicted to worsen the CS. In addition, we identified the negative regulation of female hormones among pathways potentially aggravating the CS, which helps to understand the gender-related differences in COVID-19 mortality. Regarding drugs potentially counteracting the CS, we identified glucocorticoids as a top hit, which validates our approach as this is the primary treatment for this pathology. Interestingly, our analysis also reveals a potential effect of MEK inhibitors in reverting the COVID-19 CS, which is supported by in vitro data that confirms the anti-inflammatory properties of these compounds.
- Published
- 2022
- Full Text
- View/download PDF
7. A chemical screen for modulators of mRNA translation identifies a distinct mechanism of toxicity for sphingosine kinase inhibitors.
- Author
-
Alba Corman, Dimitris C Kanellis, Patrycja Michalska, Maria Häggblad, Vanesa Lafarga, Jiri Bartek, Jordi Carreras-Puigvert, and Oscar Fernandez-Capetillo
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We here conducted an image-based chemical screen to evaluate how medically approved drugs, as well as drugs that are currently under development, influence overall translation levels. None of the compounds up-regulated translation, which could be due to the screen being performed in cancer cells grown in full media where translation is already present at very high levels. Regarding translation down-regulators, and consistent with current knowledge, inhibitors of the mechanistic target of rapamycin (mTOR) signaling pathway were the most represented class. In addition, we identified that inhibitors of sphingosine kinases (SPHKs) also reduce mRNA translation levels independently of mTOR. Mechanistically, this is explained by an effect of the compounds on the membranes of the endoplasmic reticulum (ER), which activates the integrated stress response (ISR) and contributes to the toxicity of SPHK inhibitors. Surprisingly, the toxicity and activation of the ISR triggered by 2 independent SPHK inhibitors, SKI-II and ABC294640, the latter in clinical trials, are also observed in cells lacking SPHK1 and SPHK2. In summary, our study provides a useful resource on the effects of medically used drugs on translation, identified compounds capable of reducing translation independently of mTOR and has revealed that the cytotoxic properties of SPHK inhibitors being developed as anticancer agents are independent of SPHKs.
- Published
- 2021
- Full Text
- View/download PDF
8. The RNA Polymerase II Factor RPAP1 Is Critical for Mediator-Driven Transcription and Cell Identity
- Author
-
Cian J. Lynch, Raquel Bernad, Isabel Calvo, Sandrina Nóbrega-Pereira, Sergio Ruiz, Nuria Ibarz, Ana Martinez-Val, Osvaldo Graña-Castro, Gonzalo Gómez-López, Eduardo Andrés-León, Vladimir Espinosa Angarica, Antonio del Sol, Sagrario Ortega, Oscar Fernandez-Capetillo, Enrique Rojo, Javier Munoz, and Manuel Serrano
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The RNA polymerase II-associated protein 1 (RPAP1) is conserved across metazoa and required for stem cell differentiation in plants; however, very little is known about its mechanism of action or its role in mammalian cells. Here, we report that RPAP1 is essential for the expression of cell identity genes and for cell viability. Depletion of RPAP1 triggers cell de-differentiation, facilitates reprogramming toward pluripotency, and impairs differentiation. Mechanistically, we show that RPAP1 is essential for the interaction between RNA polymerase II (RNA Pol II) and Mediator, as well as for the recruitment of important regulators, such as the Mediator-specific RNA Pol II factor Gdown1 and the C-terminal domain (CTD) phosphatase RPAP2. In agreement, depletion of RPAP1 diminishes the loading of total and Ser5-phosphorylated RNA Pol II on many genes, with super-enhancer-driven genes among the most significantly downregulated. We conclude that Mediator/RPAP1/RNA Pol II is an ancient module, conserved from plants to mammals, critical for establishing and maintaining cell identity. : Lynch et al. report a regulator of RNA Pol II called RPAP1, displaying functional conservation from plants to mammals. RPAP1 is required to establish and maintain cell identity. Mechanistically, RPAP1 is critical for the Mediator-RNA Pol II interaction, thereby preserving normal transcription at enhancer-driven genes. Keywords: transcription, RNA polymerase II, Mediator, cell identity, differentiation, interactome, enhancer
- Published
- 2018
- Full Text
- View/download PDF
9. A Chemical Screen Identifies Compounds Capable of Selecting for Haploidy in Mammalian Cells
- Author
-
Teresa Olbrich, Maria Vega-Sendino, Matilde Murga, Guillermo de Carcer, Marcos Malumbres, Sagrario Ortega, Sergio Ruiz, and Oscar Fernandez-Capetillo
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The recent availability of somatic haploid cell lines has provided a unique tool for genetic studies in mammals. However, the percentage of haploid cells rapidly decreases in these cell lines, which we recently showed is due to their overgrowth by diploid cells present in the cultures. Based on this property, we have now performed a phenotypic chemical screen in human haploid HAP1 cells aiming to identify compounds that facilitate the maintenance of haploid cells. Our top hit was 10-Deacetyl-baccatin-III (DAB), a chemical precursor in the synthesis of Taxol, which selects for haploid cells in HAP1 and mouse haploid embryonic stem cultures. Interestingly, DAB also enriches for diploid cells in mixed cultures of diploid and tetraploid cells, including in the colon cancer cell line DLD-1, revealing a general strategy for selecting cells with lower ploidy in mixed populations of mammalian cells. : Cultures of haploid animal cell lines become progressively enriched in diploid cells. By conducting a chemical screen in HAP1 cells, Olbrich et al. identify compounds that facilitate the maintenance of haploid cells and a general strategy to select for cells with lower ploidy in mixed cultures of mammalian cells. Keywords: chemical screen, haploidy, tetraploidy, 10-Deacetylbaccatin-III, paclitaxel, mitosis
- Published
- 2019
- Full Text
- View/download PDF
10. A Proteomic Characterization of Factors Enriched at Nascent DNA Molecules
- Author
-
Andres J. Lopez-Contreras, Isabel Ruppen, Maria Nieto-Soler, Matilde Murga, Sara Rodriguez-Acebes, Silvia Remeseiro, Sara Rodrigo-Perez, Ana M. Rojas, Juan Mendez, Javier Muñoz, and Oscar Fernandez-Capetillo
- Subjects
Biology (General) ,QH301-705.5 - Abstract
DNA replication is facilitated by multiple factors that concentrate in the vicinity of replication forks. Here, we developed an approach that combines the isolation of proteins on nascent DNA chains with mass spectrometry (iPOND-MS), allowing a comprehensive proteomic characterization of the human replisome and replisome-associated factors. In addition to known replisome components, we provide a broad list of proteins that reside in the vicinity of the replisome, some of which were not previously associated with replication. For instance, our data support a link between DNA replication and the Williams-Beuren syndrome and identify ZNF24 as a replication factor. In addition, we reveal that SUMOylation is widespread for factors that concentrate near replisomes, which contrasts with lower UQylation levels at these sites. This resource provides a panoramic view of the proteins that concentrate in the surroundings of the replisome, which should facilitate future investigations on DNA replication and genome maintenance.
- Published
- 2013
- Full Text
- View/download PDF
11. Targeting ATR and Chk1 kinases for cancer treatment: A new model for new (and old) drugs
- Author
-
Luis I. Toledo, Matilde Murga, and Oscar Fernandez‐Capetillo
- Subjects
ATR ,Chk1 ,Cancer therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Trying to kill cancer cells by generating DNA damage is by no means a new idea. Radiotherapy and genotoxic drugs are routinely used in cancer therapy. More recent developments also explored the potential of targeting the DNA damage response (DDR) in order to increase the toxicity of radio‐ and chemo‐ therapy. Chk1 inhibitors have pioneered studies in this regard. Interestingly, early studies noted that Chk1 inhibitors were particularly toxic for p53‐deficient cells. The model proposed for this observation was that this effect was due to the simultaneous abrogation of the G2 (Chk1) and G1 (p53) checkpoints. We here challenge this view, and propose a model where the toxicity of Chk1 inhibitors is rather due to the fact that these compounds generate high loads of replicative stress (RS) during S‐phase, which are further boosted by the less restrictive S‐phase entry found in p53‐deficient cells. This new model implies that the particular toxicity of Chk1 inhibitors might not be restricted to p53‐deficient cells, but could be extended to other mutations that promote a promiscuous S‐phase entry. In addition, this rationale also implies that the same effect should also be observed for other molecules that target the RS‐response (RSR), such as inhibitors of the Chk1‐activating kinase ATR.
- Published
- 2011
- Full Text
- View/download PDF
12. Histone H2A C-terminus regulates chromatin dynamics, remodeling, and histone H1 binding.
- Author
-
Christine Vogler, Claudia Huber, Tanja Waldmann, Ramona Ettig, Lora Braun, Annalisa Izzo, Sylvain Daujat, Isabelle Chassignet, Andres Joaquin Lopez-Contreras, Oscar Fernandez-Capetillo, Miroslav Dundr, Karsten Rippe, Gernot Längst, and Robert Schneider
- Subjects
Genetics ,QH426-470 - Abstract
The tails of histone proteins are central players for all chromatin-mediated processes. Whereas the N-terminal histone tails have been studied extensively, little is known about the function of the H2A C-terminus. Here, we show that the H2A C-terminal tail plays a pivotal role in regulating chromatin structure and dynamics. We find that cells expressing C-terminally truncated H2A show increased stress sensitivity. Moreover, both the complete and the partial deletion of the tail result in increased histone exchange kinetics and nucleosome mobility in vivo and in vitro. Importantly, our experiments reveal that the H2A C-terminus is required for efficient nucleosome translocation by ISWI-type chromatin remodelers and acts as a novel recognition module for linker histone H1. Thus, we suggest that the H2A C-terminal tail has a bipartite function: stabilisation of the nucleosomal core particle, as well as mediation of the protein interactions that control chromatin dynamics and conformation.
- Published
- 2010
- Full Text
- View/download PDF
13. Limited role of murine ATM in oncogene-induced senescence and p53-dependent tumor suppression.
- Author
-
Alejo Efeyan, Matilde Murga, Barbara Martinez-Pastor, Ana Ortega-Molina, Rebeca Soria, Manuel Collado, Oscar Fernandez-Capetillo, and Manuel Serrano
- Subjects
Medicine ,Science - Abstract
Recent studies in human fibroblasts have provided a new general paradigm of tumor suppression according to which oncogenic signaling produces DNA damage and this, in turn, results in ATM/p53-dependent cellular senescence. Here, we have tested this model in a variety of murine experimental systems. Overexpression of oncogenic Ras in murine fibroblasts efficiently induced senescence but this occurred in the absence of detectable DNA damage signaling, thus suggesting a fundamental difference between human and murine cells. Moreover, lung adenomas initiated by endogenous levels of oncogenic K-Ras presented abundant senescent cells, but undetectable DNA damage signaling. Accordingly, K-Ras-driven adenomas were also senescent in Atm-null mice, and the tumorigenic progression of these lesions was only modestly accelerated by Atm-deficiency. Finally, we have examined chemically-induced fibrosarcomas, which possess a persistently activated DNA damage response and are highly sensitive to the activity of p53. We found that the absence of Atm favored genomic instability in the resulting tumors, but did not affect the persistent DNA damage response and did not impair p53-dependent tumor suppression. All together, we conclude that oncogene-induced senescence in mice may occur in the absence of a detectable DNA damage response. Regarding murine Atm, our data suggest that it plays a minor role in oncogene-induced senescence or in p53-dependent tumor suppression, being its tumor suppressive activity probably limited to the maintenance of genomic stability.
- Published
- 2009
- Full Text
- View/download PDF
14. Targeting the nucleolus as a therapeutic strategy in human disease
- Author
-
Alba Corman, Oleksandra Sirozh, Vanesa Lafarga, and Oscar Fernandez-Capetillo
- Subjects
Molecular Biology ,Biochemistry - Abstract
The nucleolus is the site of ribosome biogenesis, one of the most resource-intensive processes in eukaryotic cells. Accordingly, nucleolar morphology and activity are highly responsive to growth signaling and nucleolar insults which are collectively included in the actively evolving concept of nucleolar stress. Importantly, nucleolar alterations are a prominent feature of multiple human pathologies, including cancer and neurodegeneration, as well as being associated with aging. The past decades have seen numerous attempts to isolate compounds targeting different facets of nucleolar activity. We provide an overview of therapeutic opportunities for targeting nucleoli in different pathologies and currently available therapies.
- Published
- 2023
- Full Text
- View/download PDF
15. Actionable cancer vulnerability due to translational arrest, p53 aggregation and ribosome biogenesis stress evoked by the disulfiram metabolite CuET
- Author
-
Dimitris C. Kanellis, Asimina Zisi, Zdenek Skrott, Bennie Lemmens, Jaime A. Espinoza, Martin Kosar, Andrea Björkman, Xuexin Li, Stefanos Arampatzis, Jirina Bartkova, Miguel Andújar-Sánchez, Oscar Fernandez-Capetillo, Martin Mistrik, Mikael S. Lindström, and Jiri Bartek
- Subjects
Cell Biology ,Molecular Biology - Abstract
Drug repurposing is a versatile strategy to improve current therapies. Disulfiram has long been used in the treatment of alcohol dependency and multiple clinical trials to evaluate its clinical value in oncology are ongoing. We have recently reported that the disulfiram metabolite diethyldithiocarbamate, when combined with copper (CuET), targets the NPL4 adapter of the p97VCP segregase to suppress the growth of a spectrum of cancer cell lines and xenograft models in vivo. CuET induces proteotoxic stress and genotoxic effects, however important issues concerning the full range of the CuET-evoked tumor cell phenotypes, their temporal order, and mechanistic basis have remained largely unexplored. Here, we have addressed these outstanding questions and show that in diverse human cancer cell models, CuET causes a very early translational arrest through the integrated stress response (ISR), later followed by features of nucleolar stress. Furthermore, we report that CuET entraps p53 in NPL4-rich aggregates leading to elevated p53 protein and its functional inhibition, consistent with the possibility of CuET-triggered cell death being p53-independent. Our transcriptomics profiling revealed activation of pro-survival adaptive pathways of ribosomal biogenesis (RiBi) and autophagy upon prolonged exposure to CuET, indicating potential feedback responses to CuET treatment. The latter concept was validated here by simultaneous pharmacological inhibition of RiBi and/or autophagy that further enhanced CuET’s tumor cytotoxicity, using both cell culture and zebrafish in vivo preclinical models. Overall, these findings expand the mechanistic repertoire of CuET’s anti-cancer activity, inform about the temporal order of responses and identify an unorthodox new mechanism of targeting p53. Our results are discussed in light of cancer-associated endogenous stresses as exploitable tumor vulnerabilities and may inspire future clinical applications of CuET in oncology, including combinatorial treatments and focus on potential advantages of using certain validated drug metabolites, rather than old, approved drugs with their, often complex, metabolic profiles.
- Published
- 2023
- Full Text
- View/download PDF
16. Data from p27Kip1 Stabilization Is Essential for the Maintenance of Cell Cycle Arrest in Response to DNA Damage
- Author
-
Oscar Fernandez-Capetillo, Angel R. Nebreda, Aneta Swat, Paula Gutierrez-Martinez, and Myriam Cuadrado
- Abstract
One of the current models of cancer proposes that oncogenes activate a DNA damage response (DDR), which would limit the growth of the tumor in its earliest stages. In this context, and in contrast to studies focused on the acute responses to a one-time genotoxic insult, understanding how cells respond to a persistent source of DNA damage might become critical for future studies in the field. We here report the discovery of a novel damage-responsive pathway, which involves p27Kip1 and retinoblastoma tumor suppressors and is only implemented after a persistent exposure to clastogens. In agreement with its late activation, we show that this pathway is critical for the maintenance, but not the initiation, of the cell cycle arrest triggered by DNA damage. Interestingly, this late response is independent of the canonical ataxia telangiectasia mutated–dependent and ataxia telangiectasia mutated and Rad3-related–dependent DDR but downstream of p38 mitogen-activated protein kinase. Our results might help to reconcile the oncogene-induced DNA damage model with the clinical evidence that points to non-DDR members as the most important tumor suppressors in human cancer. [Cancer Res 2009;69(22):8726–32]
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figures 1-6 from p27Kip1 Stabilization Is Essential for the Maintenance of Cell Cycle Arrest in Response to DNA Damage
- Author
-
Oscar Fernandez-Capetillo, Angel R. Nebreda, Aneta Swat, Paula Gutierrez-Martinez, and Myriam Cuadrado
- Abstract
Supplementary Figures 1-6 from p27Kip1 Stabilization Is Essential for the Maintenance of Cell Cycle Arrest in Response to DNA Damage
- Published
- 2023
- Full Text
- View/download PDF
18. The Drug Repurposing Encyclopedia (DRE): a web server for systematic drug repurposing across 20 organisms
- Author
-
Xuexin Li, Lu Pan, Laura Sanchez-Burgos, Daniela Hühn, and Oscar Fernandez-Capetillo
- Abstract
The identification of new therapeutic uses for compounds via computational or experimental approaches, which is widely known as drug repurposing, has the potential to develop novel therapies with pre-existing medicines, thereby reducing the time and costs associated with drug development. Today, several data-driven methodologies have been developed leading to databases that facilitate drug repurposing initiatives. However, no approach has systematically compared drug transcriptional profiles to those from a wide spectrum of human diseases or molecular pathways. Here, we present the Drug Repurposing Encyclopedia (DRE,https://www.drugrep.org), an interactive web server covering over 198M significant drug-signature associations across 20 organisms to allow users to carry out drug-repositioning analyses. DRE consists of 12 modules covering real-time drug-repurposing for user-provided transcriptional signatures; gene set enrichment analysis (GSEA) for all available drug transcriptomics profiles; as well as similarity analyses for provided gene sets across all database signatures. Collectively, DRE provides a one-stop comprehensive solution to help scientists interested in drug-repurposing studies.
- Published
- 2023
- Full Text
- View/download PDF
19. The anti-leprosy drug clofazimine reduces polyQ toxicity through activation of PPARγ
- Author
-
Xuexin Li, Ivo Hernandez, Maria Häggblad, Louise Lidemalm, Lars Brautigam, Jose J. Lucas, Jordi Carreras-Puigvert, Daniela Hühn, and Oscar Fernandez-Capetillo
- Abstract
SUMMARYPolyQ diseases are autosomal dominant neurodegenerative disorders caused by the expansion of CAG repeats. While of slow progression, these diseases are ultimately fatal and lack effective therapies. Here, we present our results from a High-Throughput chemical screen oriented to find drugs that lower the toxicity of a protein containing the first exon from the Huntington’s disease protein huntingtin (HTT) harboring 94 glutamines (Htt-Q94). Our screening identified the anti-leprosy drug clofazimine as a hit, which was subsequently validated in several in vitro models as well as in a zebrafish model of polyQ toxicity. Computational analyses of transcriptional signatures, together with molecular modeling and biochemical assays revealed that clofazimine is an agonist of the peroxisome proliferator activated receptor gamma (PPARγ), previously suggested as a potential therapy for HD by stimulating mitochondrial biogenesis. Accordingly, clofazimine rescued the mitochondrial dysfunction triggered by Htt-Q94expression. Together, our results support the potential of clofazimine repurposing for the treatment of PolyQ diseases.
- Published
- 2023
- Full Text
- View/download PDF
20. Prolonged estrogen deprivation triggers a broad immunosuppressive phenotype in breast cancer cells
- Author
-
Maria Häggblad, Silvana Mouron, Bartlomiej Porebski, Catherine Hansel, Louise Lidemalm, Pablo Martí-Rodrigo, Daniela Hühn, Jordi Carreras-Puigvert, Oscar Fernandez-Capetillo, Kirsten Tschapalda, Miguel Quintela-Fandino, Karolinska Institutet, Cancerfonden Foundation, Swedish Research Council, Instituto de Salud Carlos III, Unión Europea. Comisión Europea, and Comunidad de Madrid (España)
- Subjects
PD-L1 ,Cancer Research ,medicine.medical_treatment ,Estrogen receptor ,Breast Neoplasms ,B7-H1 Antigen ,Breast cancer ,Immune system ,breast cancer ,INFLAMMATION ,Cell Line, Tumor ,Genetics ,Medicine ,Humans ,IMMUNOTHERAPY ,Research Articles ,RC254-282 ,Cancer och onkologi ,business.industry ,Estrogen Antagonists ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Estrogens ,General Medicine ,Immunotherapy ,medicine.disease ,Antiestrogen ,HLA ,Cell killing ,Phenotype ,Oncology ,inflammation ,PD‐L1 ,Cancer and Oncology ,Cancer research ,Molecular Medicine ,Female ,immunotherapy ,BREAST-CANCER PATIENTS ,business ,Janus kinase ,Estrogen receptor alpha ,Research Article ,estrogen receptor - Abstract
Among others, expression levels of programmed cell death 1 ligand 1 (PD‐L1) have been explored as biomarkers of the response to immune checkpoint inhibitors in cancer therapy. Here, we present the results of a chemical screen that interrogated how medically approved drugs influence PD‐L1 expression. As expected, corticosteroids and inhibitors of Janus kinases were among the top PD‐L1 downregulators. In addition, we identified that PD‐L1 expression is induced by antiestrogenic compounds. Transcriptomic analyses indicate that chronic estrogen receptor alpha (ERα) inhibition triggers a broad immunosuppressive program in ER‐positive breast cancer cells, which is subsequent to their growth arrest and involves the activation of multiple immune checkpoints together with the silencing of the antigen‐presenting machinery. Accordingly, estrogen‐deprived MCF7 cells are resistant to T‐cell‐mediated cell killing, in a manner that is independent of PD‐L1, but which is reverted by estradiol. Our study reveals that while antiestrogen therapies efficiently limit the growth of ER‐positive breast cancer cells, they concomitantly trigger a transcriptional program that favors their immune evasion., Under prolonged hormone therapy, ER+ breast cancer cells activate an inflammatory transcriptional program, which includes a generalized upregulation of immune checkpoint mediators together with the downregulation of the antigen‐presenting machinery. These findings reveal that, while hormone therapies efficiently arrest the growth of ER+ breast cancer cells, they also promote a phenotype switch that favors their immune evasion.
- Published
- 2022
21. Basic Concepts and Emergent Disease Mechanisms of Amyotrophic Lateral Sclerosis
- Author
-
Oleksandra Sirozh, Anabel Saez-Mas, Vanesa Lafarga, and Oscar Fernandez-Capetillo
- Published
- 2023
- Full Text
- View/download PDF
22. A Multiparametric and High-Throughput Platform for Host-Virus Binding Screens
- Author
-
Jan Schlegel, Bartlomiej Porebski, Luca Andronico, Leo Hanke, Steven Edwards, Hjalmar Brismar, Benjamin Murrell, Gerald McInerney, Oscar Fernandez-Capetillo, and Erdinc Sezgin
- Abstract
Speed is key during infectious disease outbreaks. It is essential, for example, to identify critical host binding factors to the pathogens as fast as possible. The complexity of host plasma membrane is often a limiting factor hindering fast and accurate determination of host binding factors as well as high-throughput screening for neutralizing antimicrobial drug targets. Here we describe a multi-parametric and high-throughput platform tackling this bottleneck and enabling fast screens for host binding factors as well as new antiviral drug targets. The sensitivity and robustness of our platform was validated by blocking SARS-CoV-2 spike particles with nanobodies and IgGs from human serum samples.TeaserA fast screening platform tackling host-pathogen interactions.
- Published
- 2022
- Full Text
- View/download PDF
23. I11 High throughput screening reveal drugs to rescue polyQ induced toxicity in Huntington’s disease cell and animal models
- Author
-
Xuexin Li, Maria Häggblad, Lars Bräutigam, Jordi Carreras-Puigvert, Daniela Hühn, and Oscar Fernandez-Capetillo
- Published
- 2022
- Full Text
- View/download PDF
24. SETD8 inhibition targets cancer cells with increased rates of ribosome biogenesis
- Author
-
Matilde Murga, Gema Lopez-Pernas, Robert Soliva, Elena Fueyo-Marcos, Corina Amor, Ignacio Faustino, Marina Serna, Alicia G. Serrano, Lucía Díaz, Sonia Martínez, Carmen Blanco-Aparicio, Marta Elena Antón, Brinton Seashore-Ludlow, Joaquín Pastor, Rozbeh Jafari, Miguel Lafarga, Oscar Llorca, Modesto Orozco, and Oscar Fernández-Capetillo
- Subjects
Cytology ,QH573-671 - Abstract
Abstract SETD8 is a methyltransferase that is overexpressed in several cancers, which monomethylates H4K20 as well as other non-histone targets such as PCNA or p53. We here report novel SETD8 inhibitors, which were discovered while trying to identify chemicals that prevent 53BP1 foci formation, an event mediated by H4K20 methylation. Consistent with previous reports, SETD8 inhibitors induce p53 expression, although they are equally toxic for p53 proficient or deficient cells. Thermal stability proteomics revealed that the compounds had a particular impact on nucleoli, which was confirmed by fluorescent and electron microscopy. Similarly, Setd8 deletion generated nucleolar stress and impaired ribosome biogenesis, supporting that this was an on-target effect of SETD8 inhibitors. Furthermore, a genome-wide CRISPR screen identified an enrichment of nucleolar factors among those modulating the toxicity of SETD8 inhibitors. Accordingly, the toxicity of SETD8 inhibition correlated with MYC or mTOR activity, key regulators of ribosome biogenesis. Together, our study provides a new class of SETD8 inhibitors and a novel biomarker to identify tumors most likely to respond to this therapy.
- Published
- 2024
- Full Text
- View/download PDF
25. Supraphysiological protection from replication stress does not extend mammalian lifespan
- Author
-
Eliene Albers, Alexandra Avram, Andrés J. López-Contreras, Mauro Sbroggiò, and Oscar Fernandez-Capetillo
- Subjects
Genome instability ,Genetically modified mouse ,DNA Replication ,Aging ,medicine.medical_specialty ,Ribonucleoside Diphosphate Reductase ,DNA damage ,replication stress ,Longevity ,Context (language use) ,Endogeny ,Mice, Transgenic ,Biology ,Mouse models ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,cancer ,Animals ,mouse models ,Gene ,Cancer ,030304 developmental biology ,0303 health sciences ,Progeria ,aging ,Replication stress ,Cell Biology ,medicine.disease ,Accelerated aging ,Endocrinology ,030220 oncology & carcinogenesis ,Checkpoint Kinase 1 ,Priority Research Paper - Abstract
Replication Stress (RS) is a type of DNA damage generated at the replication fork, characterized by single-stranded DNA (ssDNA) accumulation, and which can be caused by a variety of factors. Previous studies have reported elevated RS levels in aged cells. In addition, mouse models with a deficient RS response show accelerated aging. However, the relevance of endogenous or physiological RS, compared to other sources of genomic instability, for the normal onset of aging is unknown. We have performed long term survival studies of transgenic mice with extra copies of the Chk1 and/or Rrm2 genes, which we previously showed extend the lifespan of a progeroid ATR-hypomorphic model suffering from high levels of RS. In contrast to their effect in the context of progeria, the lifespan of Chk1, Rrm2 and Chk1/Rrm2 transgenic mice was similar to WT littermates in physiological settings. Most mice studied died due to tumors -mainly lymphomas-irrespective of their genetic background. Interestingly, a slightly higher percentage of transgenic mice developed tumors compared to WT mice. Our results indicate that supraphysiological protection from RS does not extend lifespan, indicating that RS may not be a relevant source of genomic instability on the onset of “normal” aging.
- Published
- 2020
- Full Text
- View/download PDF
26. Chemical and genetic screens identify new regulators of tetracycline-inducible gene expression system in mammalian cells
- Author
-
Valeria Colicchia, Maria Häggblad, Oleksandra Sirozh, Bartlomiej Porebski, Mirela Balan, Louise Lidemalm, Jordi Carreras-Puigvert, Daniela Hühn, and Oscar Fernandez-Capetillo
- Abstract
The tetracycline repressor (tetR)-regulated system is a widely used tool to specifically control gene expression in mammalian cells. Based on this system, we generated a human osteosarcoma cell line which allows for inducible expression of an EGFP-fusion of the TAR DNA-binding protein 43 (TDP-43), which has been linked to neurodegenerative diseases. Consistent with previous findings, TDP-43 overexpression led to the accumulation of aggregates and limited the viability of U2OS. Using this inducible system, we conducted a chemical screen with a library that included FDA-approved drugs. While the primary screen identified several compounds that prevented TDP-43 toxicity, further experiments revealed that these chemicals abrogated doxycyclinedependent TDP-43 expression. This antagonistic effect was observed with both doxycycline and tetracycline, and in several Tet-On cell lines expressing different genes, confirming the general effect of these compounds as inhibitors of the tetR system. Using the same cell line, a genome-wide CRISPR/Cas9 screen identified epigenetic regulators such as the G9a methyltransferase or TRIM28 as potential modifiers of TDP-43 toxicity. Yet again, further experiments revealed that G9a inhibition or TRIM28 loss prevented doxycycline-dependent expression of TDP-43. Together, these results suggest that none of the medically approved drugs significantly mitigates TDP-43 toxicity, identify new chemical and genetic regulators of the tetR system, and raise awareness on the limitations of this approach to conduct chemical or genetic screenings in mammalian cells.
- Published
- 2022
- Full Text
- View/download PDF
27. Activation of the Integrated Stress Response overcomes multidrug resistance in FBXW7-deficient cells
- Author
-
Laura Sanchez-Burgos, Belén Navarro-González, Santiago García-Martín, Héctor Tejero, Marta Elena Antón, Fátima Al-Shahrour, and Oscar Fernandez-Capetillo
- Abstract
SUMMARYFBXW7 is one of the most frequently mutated tumor suppressors, the deficiency of which has been associated with resistance to some anticancer therapies. Through bioinformatic analyses and genome-wide CRISPR screens, we here reveal that FBXW7 deficiency leads to multidrug resistance (MDR), to a bigger extent than well-established MDR-drivers such as overexpression of the drug-efflux pump ABCB1. Proteomic data from FBXW7-deficient cancer cells identify the upregulation of mitochondrial function as a hallmark of FBXW7 deficiency, which has been previously linked to an increased resistance to chemotherapy. Accordingly, genetic or chemical targeting of mitochondria is preferentially toxic for FBXW7-deficient cells in vitro and in vivo. Mechanistically, we show that the toxicity associated with therapies that target mitochondrial translation such as the antibiotic tigecycline relates to the activation of the Integrated Stress Response (ISR). Furthermore, while searching for additional drugs that could overcome the MDR of FBXW7-deficient cells, we found that all of them unexpectedly also activated the ISR regardless of their currently accepted mechanism of action. Together, our study reveals that one of the most frequent mutations in cancer reduces the sensitivity to the vast majority of available therapies, and identifies a general principle to overcome such resistance.
- Published
- 2022
- Full Text
- View/download PDF
28. Coordinating DNA Replication and Mitosis through Ubiquitin/SUMO and CDK1
- Author
-
Oscar Fernandez-Capetillo, Antonio Galarreta, Emilio Lecona, Pablo Valledor, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), European Research Council, Fundación Ramón Areces, La Caixa, Consejo Superior de Investigaciones Científicas (España), and European Commission
- Subjects
CDK1 ,DNA replication initiation ,DNA damage ,DNA repair ,QH301-705.5 ,SUMO protein ,Review ,Biology ,DNA replication ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,0302 clinical medicine ,CDC2 Protein Kinase ,ubiquitin ,Animals ,Humans ,Physical and Theoretical Chemistry ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,030304 developmental biology ,mitosis ,0303 health sciences ,Organic Chemistry ,General Medicine ,3. Good health ,Computer Science Applications ,Chromatin ,Cell biology ,Chemistry ,SUMO ,Small Ubiquitin-Related Modifier Proteins ,DNA replication termination ,USP7 ,Replisome ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
Post‐translational modification of the DNA replication machinery by ubiquitin and SUMO plays key roles in the faithful duplication of the genetic information. Among other functions, ubiquitination and SUMOylation serve as signals for the extraction of factors from chromatin by the AAA ATPase VCP. In addition to the regulation of DNA replication initiation and elongation, we now know that ubiquitination mediates the disassembly of the replisome after DNA replication termination, a process that is essential to preserve genomic stability. Here, we review the recent evidence showing how active DNA replication restricts replisome ubiquitination to prevent the premature disassembly of the DNA replication machinery. Ubiquitination also mediates the removal of the replisome to allow DNA repair. Further, we discuss the interplay between ubiquitinmediated replisome disassembly and the activation of CDK1 that is required to set up the transition from the S phase to mitosis. We propose the existence of a ubiquitin–CDK1 relay, where the disassembly of terminated replisomes increases CDK1 activity that, in turn, favors the ubiquitination and disassembly of more replisomes. This model has important implications for the mechanism of action of cancer therapies that induce the untimely activation of CDK1, thereby triggering premature replisome disassembly and DNA damage., Ramón y Cajal Fellowship from MINECO (RYC-2016-20705), co-funded by European Regional Development Funds (FEDER) to Emilio Lecona; by grants from the Spanish Ministry of Science, Innovation and Universities (RTI2018-102204-B-I00, co-financed with European FEDER funds) and the European Research Council (ERC-617840) to Oscar Fernandez-Capetillo; fellowships from Fundacion Ramon Areces-UAM and LaCaixa Foundation to Pablo Valledor (LCF/BQ/ES18/11670008)
- Published
- 2021
29. Author response for 'Prolonged estrogen deprivation triggers a broad immunosuppressive phenotype in breast cancer cells'
- Author
-
Jordi Carreras-Puigvert, Pablo Marti-Rodrigo, Daniela Hühn, Silvana Mouron, Miguel Quintela-Fandino, Bartlomiej Porebski, Oscar Fernandez-Capetillo, Catherine Hansel, Maria Häggblad, Louise Lidemalm, and Kirsten Tschapalda
- Subjects
business.industry ,Cancer research ,Medicine ,Breast cancer cells ,Estrogen deprivation ,business ,Phenotype - Published
- 2021
- Full Text
- View/download PDF
30. Review for 'Targeting mitochondrial respiration and the BCL2 family in high‐grade MYC‐associated B‐cell lymphoma'
- Author
-
Oscar Fernandez-Capetillo
- Subjects
Cancer research ,medicine ,Biology ,B-cell lymphoma ,medicine.disease ,Mitochondrial respiration - Published
- 2021
- Full Text
- View/download PDF
31. Widespread displacement of DNA- and RNA-binding factors underlies toxicity of arginine-rich cell-penetrating peptides
- Author
-
Rafael Fernandez-Leiro, Irene Díaz-López, Bogdan Jovanovic, Oscar Fernandez-Capetillo, Iván Ventoso, Jasminka Boskovic, Georg Stoecklin, Oleksandra Sirozh, Eduardo Zarzuela, Antonio Galarreta, Misaru Hisaoka, Jaime Muñoz, Vanesa Lafarga, Fundación Botín, Ministerio de Ciencia, Innovación y Universidades (España), European Commission, and German Research Foundation
- Subjects
RNA Splicing ,mRNA ,Protamine ,Cell-Penetrating Peptides ,Biology ,Arginine ,General Biochemistry, Genetics and Molecular Biology ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Nucleic Acids ,Humans ,RNA, Messenger ,Spermatogenesis ,Molecular Biology ,Gene ,030304 developmental biology ,0303 health sciences ,Messenger RNA ,General Immunology and Microbiology ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,RNA ,RNA-Binding Proteins ,DNA ,Articles ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Histone ,chemistry ,Arginine-rich peptides ,RNA splicing ,biology.protein ,Nucleic acid ,ALS ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Due to their capability to transport chemicals or proteins into target cells, cell-penetrating peptides (CPPs) are being developed as therapy delivery tools. However, and despite their interesting properties, arginine-rich CPPs often show toxicity for reasons that remain poorly understood. Using a (PR)n dipeptide repeat that has been linked to amyotrophic lateral sclerosis (ALS) as a model of an arginine-rich CPP, we here show that the presence of (PR)n leads to a generalized displacement of RNA- and DNA-binding proteins from chromatin and mRNA. Accordingly, any reaction involving nucleic acids, such as RNA transcription, translation, splicing and degradation, or DNA replication and repair, is impaired by the presence of the CPPs. Interestingly, the effects of (PR)n are fully mimicked by protamine, a small arginine-rich protein that displaces histones from chromatin during spermatogenesis. We propose that widespread coating of nucleic acids and consequent displacement of RNA- and DNA-binding factors from chromatin and mRNA accounts for the toxicity of arginine-rich CPPs, including those that have been recently associated with the onset of ALS., Fundación Botín, by Banco Santander through its Santander Universities Global Division and by grants from the Spanish Ministry of Science, Innovation and Universities (RTI2018-102204-B-I00, co-financed with European FEDER funds) and the European Research Council (ERC-617840) to OF; DKFZ NCT3.0 Integrative Project in Cancer Research grant (NCT3.0_2015.54 DysregPT) and SFB 1036/TP07 from the Deutsche Forschungsgemeinschaft to G.S.
- Published
- 2021
32. Distinct roles for PARP-1 and PARP-2 in c-Myc-driven B-cell lymphoma in mice
- Author
-
Nura Lutfi, Julian E. Sale, Matilde Murga, Lluis Colomo, Talia Velasco-Hernandez, Carlos Martinez, Ramon Gimeno, Françoise Dantzer, Gaël Roué, Anna Bigas, José Yélamos, Coral Ampurdanés, Guillaume Guilbaud, Oscar Fernandez-Capetillo, Violeta García-Hernández, Juan Martín-Caballero, Pilar Navarro, Sarah Bonnin, Pablo Menendez, Miguel Galindo-Campos, Biotechnologie et signalisation cellulaire (BSC), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Institut de recherche de l'Ecole de biotechnologie de Strasbourg (IREBS), Ministerio de Industria y Competitividad (España), Ministerio de Ciencia e Innovación (España), Fundación Científica Asociación Española Contra el Cáncer, Medical Research Council (UK), Laboratory of Excellence of Biology for Psychiatry (France), Université de Strasbourg, Centre National de la Recherche Scientifique (France), Instituto de Salud Carlos III, European Commission, Generalitat de Catalunya, Centres de Recerca de Catalunya, Josep Carreras Leukemia Foundation, Fundación Unoentrecienmil, and Marie Curie International Incoming Fellowship
- Subjects
Programmed cell death ,Lymphoma, B-Cell ,Carcinogenesis ,DNA damage ,Poly ADP ribose polymerase ,Immunology ,Poly (ADP-Ribose) Polymerase-1 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Biology ,Biochemistry ,Proinflammatory cytokine ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,B-cell lymphoma ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Proto-Oncogene Proteins c-myc / genetics ,Oncogene ,Poly (ADP-Ribose) Polymerase-1 / genetics ,Cancer ,Carcinogenesis / genetics ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Cell Biology ,Hematology ,medicine.disease ,3. Good health ,Lymphoma ,Gene Expression Regulation, Neoplastic ,Gene Expression Regulation ,Neoplastic Lymphoma ,030220 oncology & carcinogenesis ,Cancer research ,B-Cell / genetics ,Poly(ADP-ribose) Polymerases ,Gene Deletion ,Poly(ADP-ribose) Polymerases / genetics ,DNA Damage - Abstract
Dysregulation of the c-Myc oncogene occurs in a wide variety of hematologic malignancies, and its overexpression has been linked with aggressive tumor progression. Here, we show that poly (ADP-ribose) polymerase 1 (PARP-1) and PARP-2 exert opposing influences on progression of c-Myc-driven B-cell lymphoma. PARP-1 and PARP-2 catalyze the synthesis and transfer of ADP-ribose units onto amino acid residues of acceptor proteins in response to DNA strand breaks, playing a central role in the response to DNA damage. Accordingly, PARP inhibitors have emerged as promising new cancer therapeutics. However, the inhibitors currently available for clinical use are not able to discriminate between individual PARP proteins. We found that genetic deletion of PARP-2 prevents c-Myc-driven B-cell lymphoma, whereas PARP-1 deficiency accelerates lymphomagenesis in the E¿-Myc mouse model of aggressive B-cell lymphoma. Loss of PARP-2 aggravates replication stress in preleukemic E¿-Myc B cells, resulting in accumulation of DNA damage and concomitant cell death that restricts the c-Myc-driven expansion of B cells, thereby providing protection against B-cell lymphoma. In contrast, PARP-1 deficiency induces a proinflammatory response and an increase in regulatory T cells, likely contributing to immune escape of B-cell lymphoma, resulting in an acceleration of lymphomagenesis. These findings pinpoint specific functions for PARP-1 and PARP-2 in c-Myc-driven lymphomagenesis with antagonistic consequences that may help inform the design of new PARP-centered therapeutic strategies, with selective PARP-2 inhibition potentially representing a new therapeutic approach for the treatment of c-Myc-driven tumors., The J.Y. laboratory is funded by the Spanish Ministerio de Economía, Industria y Competitividad (grant SAF2017-83565-R), Spanish Ministerio de Ciencia e Innovación (grant PID2020-112526RB-I00), and Fundación Científica de la Asociación Española Contra el Cáncer (grant PROYEI6018YÉLA). Work in the J.E.S. laboratory is supported by a core grant to the Laboratory of Molecular Biology from the Medical Research Council (U105178808). The F.D. laboratory is supported by a Laboratory of Excellence grant (ANR-10-LABX-0034_Medalis) to Strasbourg University, Centre National de la Recherche Scientifique. The P.N. laboratory is supported by grants from the Spanish Ministry of Economy and Competitiveness/Instituto de Salud Carlos III–Fondo Europeo de Desarrollo Regional (FEDER; PI17/00199 and PI20/00625) and the Generalitat de Catalunya (2017-SGR-225). The P.M. laboratory acknowledges support from Centres de Recerca de Catalunya/Generalitat de Catalunya and Fundació Josep Carreras-Obra Social la Caixa for core support, the Spanish Ministry of Economy and Competitiveness (grant SAF-2019-108160-R), the Fundación Uno entre Cienmil, the Obra Social La Caixa (grant LCF/PR/HR19/52160011), and the German Josep Carreras Leukamie Stiftung. Work at the G.R. and P.M. laboratories are cofinanced by the European Regional Development Fund through the Interreg V-A Spain-France-Andorra Program (project PROTEOblood; grant EFA360/19). The O.F.-C. laboratory is funded by grants from the Spanish Ministry of Science, Innovation and Universities (RTI2018-102204-B-I00; cofinanced with European FEDER funds) and the European Research Council (ERC-617840). T.V.-H. was supported by a Marie Sklodowska Curie fellowship (GA792923). The A.B. laboratory is supported by the Spanish Ministry of Economy and Competitiveness (grant PID2019-104695RB-I00).
- Published
- 2021
- Full Text
- View/download PDF
33. USP7 limits CDK1 activity throughout the cell cycle
- Author
-
Javier Munoz, Antonio Galarreta, Patricia Ubieto-Capella, Marcos Malumbres, Eduardo Zarzuela, Emilio Lecona, Oscar Fernandez-Capetillo, Pablo Valledor, Vanesa Lafarga, Ministerio de Ciencia, Innovación y Universidades (España), European Commission, and Ministerio de Economía y Competitividad (España)
- Subjects
DNA damage ,Phosphatase ,Biology ,environment and public health ,General Biochemistry, Genetics and Molecular Biology ,Ubiquitin-Specific Peptidase 7 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,CDC2 Protein Kinase ,Animals ,Humans ,Protease Inhibitors ,News & Views ,Protein Phosphatase 2 ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Cyclin-dependent kinase 1 ,General Immunology and Microbiology ,General Neuroscience ,Cell Cycle ,DNA replication ,Protein phosphatase 2 ,Cell cycle ,HCT116 Cells ,Cell biology ,Protein Transport ,enzymes and coenzymes (carbohydrates) ,Cytoplasm ,NIH 3T3 Cells ,Phosphorylation ,030217 neurology & neurosurgery ,DNA Damage - Abstract
Chemical inhibitors of the deubiquitinase USP7 are currently being developed as anticancer agents based on their capacity to stabilize P53. Regardless of this activity, USP7 inhibitors also generate DNA damage in a p53-independent manner. However, the mechanism of this genotoxicity and its contribution to the anticancer effects of USP7 inhibitors are still under debate. Here we show that, surprisingly, even if USP7 inhibitors stop DNA replication, they also induce a widespread activation of CDK1 throughout the cell cycle, which leads to DNA damage and is toxic for mammalian cells. In addition, USP7 interacts with the phosphatase PP2A and supports its active localization in the cytoplasm. Accordingly, inhibition of USP7 or PP2A triggers very similar changes of the phosphoproteome, including a widespread increase in the phosphorylation of CDK1 targets. Importantly, the toxicity of USP7 inhibitors is alleviated by lowering CDK1 activity or by chemical activation of PP2A. Our work reveals that USP7 limits CDK1 activity at all cell cycle stages, providing a novel mechanism that explains the toxicity of USP7 inhibitors through untimely activation of CDK1., Spanish Ministry of Science, Innovation and Universities (RTI2018-102204-B-I00, co-financed with European FEDER funds) and the European Research Council (ERC-617840) to OF; a grant from the Spanish Ministry of Science, Innovation and Universities (RTI2018-095582-B-I00, co-financed with European FEDER funds) to MM; a grant from MINECO (BFU2014-55168-JIN)
- Published
- 2021
34. An In-Silico Analysis of Drugs Potentially Modulating the Cytokine Storm Triggered by SARS-CoV-2 Infection
- Author
-
Laura Sanchez-Burgos, Gonzalo Gómez-López, Fatima Al-Shahrour, and Oscar Fernandez-Capetillo
- Subjects
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,In silico ,medicine ,Biology ,Cytokine storm ,medicine.disease ,Virology - Abstract
The ongoing COVID-19 pandemic is one of the biggest health and societal challenges of the recent decades. Among the causes of mortality triggered by SARS-CoV-2 infection, the presence of an inflammatory "cytokine storm" (CS) at later stages of the disease has been found to play a determinant role. Here, we used available transcriptomic data from the bronchoalveolar lavage fluid (BALF) of COVID-19 patients suffering from a CS to obtain gene-signatures associated to this pathological process. Using these signatures, we interrogated the Connectivity MAP (CMap) dataset that contains the effects of over 5,000 small molecules on the transcriptome of human cancer cell lines, and looked for molecules which effects on transcription mimic or oppose those associated to the CS. Consistent with their medical use, this analysis found a significant enrichment of glucocorticoids or inhibitors of the Janus Kinases (JAK) as drugs that could revert the CS. On the other hand, molecules that potentiate the immune response such as PKC activators are predicted to worsen the CS. Besides these expected findings, our analysis also reveals a potential effect of the antibiotic doxycycline or MAPK/RAF/MEK inhibitors in reverting the CS, or of topoisomerase inhibitors and the anti-alcohol abuse drug disulfiram in potentiating its effects. Finally, our analyses support that the gender-related differences in the severity of COVID-19 are related to the anti-inflammatory properties of female hormones. While acknowledging that this is an analysis based on limited available data, we decided to share it as a resource that might help others in the selection of drugs that could be tested in the context of experimental models of CS triggered by viral infections.
- Published
- 2020
- Full Text
- View/download PDF
35. A chemical screen identifies a link between lipid metabolism and mRNA translation
- Author
-
Jiri Bartek, Alba Corman, Jordi Carreras-Puigvert, Maria Häggblad, Vanesa Lafarga, Dimitris C. Kanellis, and Oscar Fernandez-Capetillo
- Subjects
Sphingosine ,Kinase ,Endoplasmic reticulum ,MTOR signaling pathway ,Lipid metabolism ,Translation (biology) ,Biology ,law.invention ,Cell biology ,chemistry.chemical_compound ,chemistry ,law ,Suppressor ,Integrated stress response ,PI3K/AKT/mTOR pathway - Abstract
mRNA translation is one of the most energy-demanding processes for living cells, alterations of which have been frequently documented in human disease. Using recently developed technologies that enable image-based quantitation of overall translation levels, we here conducted a chemical screen to evaluate how medically approved drugs, as well as drugs that are currently under development, influence overall translation levels. Consistent with current knowledge, inhibitors of the mTOR signaling pathway were the most represented class among translation suppresors. In addition, we identified that inhibitors of sphingosine kinases (SPHKs) also reduce mRNA translation levels independently of mTOR. Mechanistically this is explained by an effect of the compounds on the membranes of the endoplasmic reticulum, which activates the integrated stress response (ISR). Accordingly, the impact of SPHK inhibitors on translation is alleviated by the concomitant inhibition of ISR kinases. On the other hand, and despite the large number of molecules tested, our study failed to identify chemicals capable of substantially increasing mRNA translation, raising doubts on to what extent translation can be supra-physiologically stimulated in mammalian cells. In summary, our study provides the first comprehensive characterization of the effect of known drugs on protein translation and has helped to unravel a new link between lipid metabolism and mRNA translation in human cells.
- Published
- 2020
- Full Text
- View/download PDF
36. Decision letter: Parallel CRISPR-Cas9 screens clarify impacts of p53 on screen performance
- Author
-
Oscar Fernandez-Capetillo and Andrea Ventura
- Subjects
Computer science ,CRISPR ,Computational biology - Published
- 2020
- Full Text
- View/download PDF
37. ATR expands embryonic stem cell fate potential in response to replication stress
- Author
-
Sara Samadi Shams, Endre Sebestyén, Sina Atashpaz, Valeria Cancila, Negar Arghavanifard, Eliene Albers, Javier Martin Gonzalez, Oscar Fernandez-Capetillo, Giovanni Faga, Angela Bachi, Vincenzo Costanzo, Elisa Allievi, Francesco Ferrari, Andrea Gnocchi, Paolo Soffientini, Simone Minardi, Claudio Tripodo, Andrés J. López-Contreras, Atashpaz S., Shams S.S., Gonzalez J.M., Sebestyen E., Arghavanifard N., Gnocchi A., Albers E., Minardi S., Faga G., Soffientini P., Allievi E., Cancila V., Bachi A., Fernandez-Capetillo O., Tripodo C., Ferrari F., Lopez-Contreras A.J., Costanzo V., Italian Association for Cancer Research, Giovanni Armenise-Harvard Foundation, European Research Council, Danish Cancer Society, Det Frie Forskningsrad (DFF), Danish National Research Foundation, Associazione Italiana per la Ricerca sul Cancro (AIRC), European Research Council (ERC), and Danmarks Grundforskningsfond
- Subjects
0301 basic medicine ,Endogeny ,Ataxia Telangiectasia Mutated Proteins ,Mice ,0302 clinical medicine ,Tandem Mass Spectrometry ,Transcription (biology) ,GENE ATR ,cell biology ,Cloning, Molecular ,Biology (General) ,Cells, Cultured ,0303 health sciences ,General Neuroscience ,Totipotent ,Cell Differentiation ,Embryo ,General Medicine ,Cell biology ,Medicine ,biological phenomena, cell phenomena, and immunity ,Research Article ,QH301-705.5 ,replication stress ,DNA damage ,Science ,Settore MED/08 - Anatomia Patologica ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Animals ,RNA, Messenger ,Gene ,Embryonic Stem Cells ,mouse ,Cell Proliferation ,030304 developmental biology ,Messenger RNA ,General Immunology and Microbiology ,Chimera ,Sequence Analysis, RNA ,Embryogenesis ,TELOMERE ELONGATION ,EPIGENETIC RESTRICTION ,embryonic stem cell ,Embryonic stem cell ,ATR ,030104 developmental biology ,Gene Expression Regulation ,DNA-DAMAGE ,Checkpoint Kinase 1 ,GENOMIC STABILITY ,030217 neurology & neurosurgery ,Chromatography, Liquid ,DNA Damage - Abstract
Fondazione Italiana per la Ricerca sul Cancro FIRC 18112 Sina Atashpaz.Fondazione Umberto Veronesi Sina Atashpaz Associazione Italiana per la Ricerca sul Cancro AIRC 5xmille METAMECH program Vincenzo Costanzo Giovanni Armenise-Harvard Foundation Vincenzo Costanzo European Research Council Consolidator grant 614541 Vincenzo Costanzo Associazione Italiana per la Ricerca sul Cancro Fellowship 23961 Negar ArghavanifarDanish Cancer Society KBVU-2014 Andres Joaquin Lopez-Contreras Danish Council for Independent Research Sapere Aude, DFF Starting Grant 2014 Andres Joaquin Lopez-Contreras European Research Council ERC-2015-STG-679068 Andres Joaquin Lopez-Contreras Danish National Research Foundation DNRF115 Andres Joaquin Lopez-Contreras The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication. Unrepaired DNA damage during embryonic development can be potentially inherited by a large population of cells. However, the quality control mechanisms that minimize the contribution of damaged cells to developing embryos remain poorly understood. Here, we uncovered an ATR- and CHK1-mediated transcriptional response to replication stress (RS) in mouse embryonic stem cells (ESCs) that induces genes expressed in totipotent two-cell (2C) stage embryos and 2C-like cells. This response is mediated by Dux, a multicopy retrogene defining the cleavage-specific transcriptional program in placental mammals. In response to RS, DUX triggers the transcription of 2C-like markers such as murine endogenous retrovirus-like elements (MERVL) and Zscan4. This response can also be elicited by ETAA1-mediated ATR activation in the absence of RS. ATR-mediated activation of DUX requires GRSF1-dependent post-transcriptional regulation of Dux mRNA. Strikingly, activation of ATR expands ESCs fate potential by extending their contribution to both embryonic and extra-embryonic tissues. These findings define a novel ATR dependent pathway involved in maintaining genome stability in developing embryos by controlling ESCs fate in response to RS. Sí
- Published
- 2020
- Full Text
- View/download PDF
38. ATR is required to complete meiotic recombination in mice
- Author
-
Scott Keeney, Ana Martínez-Marchal, Cristina Rojas, Maria Jasin, Julian Lange, Andros Maldonado-Linares, Sarai Pacheco, Ignasi Roig, Oscar Fernandez-Capetillo, Marina Marcet-Ortega, Montserrat Garcia-Caldés, Judit Fuentes-Lazaro, Ministerio de Ciencia e Innovación (España), American Cancer Society, Howard Hughes Medical Institute, and National Institutes of Health (Estados Unidos)
- Subjects
0301 basic medicine ,Male ,RAD51 ,General Physics and Astronomy ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Genetic recombination ,0302 clinical medicine ,Spermatocytes ,Testis ,DNA Breaks, Double-Stranded ,lcsh:Science ,Homologous Recombination ,In Situ Hybridization, Fluorescence ,Genetics ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Synapsis ,Nuclear Proteins ,Cell biology ,Meiosis ,biological phenomena, cell phenomena, and immunity ,Mice, 129 Strain ,DNA damage ,Science ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Prophase ,stomatognathic system ,Homologous chromosome ,Animals ,Replication protein A ,030304 developmental biology ,fungi ,General Chemistry ,Phosphate-Binding Proteins ,Mice, Inbred C57BL ,enzymes and coenzymes (carbohydrates) ,Chromosome Pairing ,030104 developmental biology ,Checkpoint Kinase 1 ,lcsh:Q ,DMC1 ,Rad51 Recombinase ,Homologous recombination ,Ataxia telangiectasia and Rad3 related ,030217 neurology & neurosurgery - Abstract
Precise execution of recombination during meiosis is essential for forming chromosomally-balanced gametes. Meiotic recombination initiates with the formation and resection of DNA double-strand breaks (DSBs). Cellular responses to meiotic DSBs are critical for efficient repair and quality control, but molecular features of these remain poorly understood, particularly in mammals. Here we report that the DNA damage response protein kinase ATR is crucial for meiotic recombination and completion of meiotic prophase in mice. Using a hypomorphic Atr mutation and pharmacological inhibition of ATR in vivo and in cultured spermatocytes, we show that ATR, through its effector kinase CHK1, promotes efficient RAD51 and DMC1 assembly at RPA-coated resected DSB sites and establishment of interhomolog connections during meiosis. Furthermore, our findings suggest that ATR promotes local accumulation of recombination markers on unsynapsed axes during meiotic prophase to favor homologous chromosome synapsis. These data reveal that ATR plays multiple roles in mammalian meiotic recombination., ATR kinase is required for meiosis in non-mammalian model organisms. Here the authors demonstrate, using a hypomorphic Atr mutation and chemical inhibition, that ATR is also essential for male meiosis in mouse, regulating meiotic recombination and synapsis.
- Published
- 2018
39. USP7 and VCPFAF1 define the SUMO/Ubiquitin landscape at the DNA replication fork
- Author
-
Emilio Lecona, Patricia Ubieto-Capella, André Franz, Domenic Pilger, Thorsten Hoppe, Antonio Galarreta, Fabian den Brave, Pablo Valledor, Guillermo de la Vega-Barranco, Alejandro Fernández-Llorente, Vanesa Lafarga, and Oscar Fernandez-Capetillo
- Subjects
DNA Replication ,ATPase ,DUB ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cofactor ,Animals, Genetically Modified ,Evolution, Molecular ,Ubiquitin-Specific Peptidase 7 ,Ubiquitin ,Valosin Containing Protein ,ubiquitin ,Endopeptidases ,Animals ,Humans ,UBXN-3 ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,FAF1 ,Adaptor Proteins, Signal Transducing ,MATH-33 ,biology ,DNA synthesis ,Chemistry ,Ubiquitination ,DNA replication ,Sumoylation ,HCT116 Cells ,DNA Replication Fork ,biology.organism_classification ,Chromatin ,Cell biology ,CDC-48 ,SUMO ,USP7 ,MCF-7 Cells ,biology.protein ,Apoptosis Regulatory Proteins ,Carrier Proteins ,VCP ,HeLa Cells - Abstract
Summary The AAA+ ATPase VCP regulates the extraction of SUMO and ubiquitin-modified DNA replication factors from chromatin. We have previously described that active DNA synthesis is associated with a SUMO-high/ubiquitin-low environment governed by the deubiquitylase USP7. Here, we unveil a functional cooperation between USP7 and VCP in DNA replication, which is conserved from Caenorhabditis elegans to mammals. The role of VCP in chromatin is defined by its cofactor FAF1, which facilitates the extraction of SUMOylated and ubiquitylated proteins that accumulate after the block of DNA replication in the absence of USP7. The inactivation of USP7 and FAF1 is synthetically lethal both in C. elegans and mammalian cells. In addition, USP7 and VCP inhibitors display synergistic toxicity supporting a functional link between deubiquitylation and extraction of chromatin-bound proteins. Our results suggest that USP7 and VCPFAF1 facilitate DNA replication by controlling the balance of SUMO/Ubiquitin-modified DNA replication factors on chromatin., Graphical abstract, Highlights • VCP and USP7 control the SUMO/ubiquitin landscape during DNA replication • VCP is recruited by FAF1 to chromatin-associated SUMO/ubiquitylated proteins • FAF1 recognizes both SUMO and ubiquitin on target proteins • The control of DNA replication by VCPFAF1 and USP7 is evolutionarily conserved, Chromatin at DNA replication forks is SUMO-rich and ubiquitin-poor. Franz et al. report that FAF1 is a dual reader of SUMO and ubiquitin targeting VCP to extract DNA replication factors from chromatin in C. elegans and human cells. Together with USP7, VCPFAF1 controls the SUMO/ubiquitin landscape during replication.
- Published
- 2021
- Full Text
- View/download PDF
40. Estrogen deprivation triggers an immunosuppressive phenotype in breast cancer cells
- Author
-
Daniela Hühn, Pablo Martí-Rodrigo, Silvana Mouron, Catherine S. Hansel, Kirsten Tschapalda, Bartlomiej Porebski, Maria Häggblad, Louise Lidemalm, Miguel A. Quintela-Fandino, Jordi Carreras-Puigvert, and Oscar Fernandez-Capetillo
- Subjects
0303 health sciences ,medicine.drug_class ,business.industry ,Estrogen receptor ,Antiestrogen ,medicine.disease ,3. Good health ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cell killing ,Breast cancer ,Estrogen ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Hormonal therapy ,business ,skin and connective tissue diseases ,hormones, hormone substitutes, and hormone antagonists ,030304 developmental biology - Abstract
Estrogen receptor (ER)-positive breast tumors are routinely treated with estrogen-depriving therapies. Despite their effectiveness, patients often progress into a more aggressive form of the disease. Through a chemical screen oriented to identify chemicals capable of inducing the expression of the immune-checkpoint ligand PD-L1, we found antiestrogens as hits. Subsequent validations confirmed that estrogen deprivation or ERα depletion induces PD-L1 expression in ER-positive breast cancer cells, both in vitro and in vivo. Likewise, PD-L1 expression is increased in metastasis arising from breast cancer patients receiving adjuvant hormonal therapy for their local disease. Transcriptome analyses indicate that estrogen deprivation triggers a broad immunosuppressive program, not restricted to PD-L1. Accordingly, estrogen deprived MCF7 cells are resistant to T-cell mediated cell killing, in a manner that can be reverted by estradiol. Our study reveals that while antiestrogen therapies effectively limit tumor growth in ER-positive breast cancers, they also trigger a transcriptional program that favors immune evasion.
- Published
- 2019
- Full Text
- View/download PDF
41. A Chemical Screen Identifies Compounds Capable of Selecting for Haploidy in Mammalian Cells
- Author
-
Sagrario Ortega, Oscar Fernandez-Capetillo, Guillermo de Cárcer, Sergio Ruiz, Teresa Olbrich, Matilde Murga, Marcos Malumbres, Maria Vega-Sendino, Boehringer Ingelheim Fonds, Ministerio de Economía y Competitividad (España), Botín Foundation, European Research Council, Fundación Banco Santander, Asociación Española Contra el Cáncer, and European Commission
- Subjects
0301 basic medicine ,Paclitaxel ,Somatic cell ,Mitosis ,Cell Separation ,Biology ,10-Deacetylbaccatin-III ,Haploidy ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Animals ,Humans ,HAP1 cells ,lcsh:QH301-705.5 ,Embryonic Stem Cells ,Ploidies ,fungi ,Chemical screen ,Diploidy ,Phenotype ,Embryonic stem cell ,High-Throughput Screening Assays ,3. Good health ,Cell biology ,Tetraploidy ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,Cell culture ,Taxoids ,Ploidy ,030217 neurology & neurosurgery - Abstract
Summary The recent availability of somatic haploid cell lines has provided a unique tool for genetic studies in mammals. However, the percentage of haploid cells rapidly decreases in these cell lines, which we recently showed is due to their overgrowth by diploid cells present in the cultures. Based on this property, we have now performed a phenotypic chemical screen in human haploid HAP1 cells aiming to identify compounds that facilitate the maintenance of haploid cells. Our top hit was 10-Deacetyl-baccatin-III (DAB), a chemical precursor in the synthesis of Taxol, which selects for haploid cells in HAP1 and mouse haploid embryonic stem cultures. Interestingly, DAB also enriches for diploid cells in mixed cultures of diploid and tetraploid cells, including in the colon cancer cell line DLD-1, revealing a general strategy for selecting cells with lower ploidy in mixed populations of mammalian cells., Graphical Abstract, Highlights • Mammalian haploid cell cultures become progressively enriched in diploid cells • DAB, a precursor of Taxol, facilitates the maintenance of haploidy • DAB selects for cells with lower ploidy in mixed cultures of mammalian cells • Statins accelerate the gradual loss of haploid cells in culture, Cultures of haploid animal cell lines become progressively enriched in diploid cells. By conducting a chemical screen in HAP1 cells, Olbrich et al. identify compounds that facilitate the maintenance of haploid cells and a general strategy to select for cells with lower ploidy in mixed cultures of mammalian cells.
- Published
- 2019
42. A SUMO and ubiquitin code coordinates protein traffic at replication factories
- Author
-
Emilio Lecona and Oscar Fernandez-Capetillo
- Subjects
0301 basic medicine ,Genetics ,biology ,Eukaryotic DNA replication ,Computational biology ,Pre-replication complex ,General Biochemistry, Genetics and Molecular Biology ,DNA replication factor CDT1 ,03 medical and health sciences ,030104 developmental biology ,Licensing factor ,Minichromosome maintenance ,Control of chromosome duplication ,biology.protein ,Replisome ,Origin recognition complex - Abstract
Post-translational modifications regulate each step of DNA replication to ensure the faithful transmission of genetic information. In this context, we recently showed that deubiquitination of SUMO2/3 and SUMOylated proteins by USP7 helps to create a SUMO-rich and ubiquitin-low environment around replisomes that is necessary to maintain the activity of replication forks and for new origin firing. We propose that a two-flag system mediates the collective concentration of factors at sites of DNA replication, whereby SUMO and Ubiquitinated-SUMO would constitute "stay" or "go" signals respectively for replisome and accessory factors. We here discuss the findings that led to this model, which have implications for the potential use of USP7 inhibitors as anticancer agents.
- Published
- 2016
- Full Text
- View/download PDF
43. POLD3 Is Haploinsufficient for DNA Replication in Mice
- Author
-
Oscar Fernandez-Capetillo, Irene Kamileri, Thanos D. Halazonetis, Emilio Lecona, Matilde Murga, Juan Méndez, Natalia Lugli, Marta Elena Anton, Sotirios K. Sotiriou, Marcos Díaz, Botín Foundation, Banco Santander, Ministerio de Economía y Competitividad (España), Fundación La Marató TV3, Howard Hughes Medical Institute, and European Research Council
- Subjects
DNA Replication ,0301 basic medicine ,DNA polymerase ,Longevity ,Eukaryotic DNA replication ,Haploinsufficiency ,Pre-replication complex ,Article ,Histones ,DNA replication factor CDT1 ,Mice ,03 medical and health sciences ,Replication factor C ,Control of chromosome duplication ,Animals ,Phosphorylation ,POLD3 Gene ,Lung ,Molecular Biology ,DNA Polymerase III ,Mice, Knockout ,Genetics ,B-Lymphocytes ,Cell Death ,030102 biochemistry & molecular biology ,biology ,Homozygote ,DNA replication ,Brain ,Gene Expression Regulation, Developmental ,Cell Biology ,Survival Analysis ,3. Good health ,030104 developmental biology ,Checkpoint Kinase 1 ,biology.protein ,DNA Damage ,Hydrocephalus - Abstract
The Pold3 gene encodes a subunit of the Polδ DNA polymerase complex. Pold3 orthologs are not essential in Saccharomyces cerevisiae or chicken DT40 cells, but the Schizosaccharomyces pombe ortholog is essential. POLD3 also has a specialized role in the repair of broken replication forks, suggesting that POLD3 activity could be particularly relevant for cancer cells enduring high levels of DNA replication stress. We report here that POLD3 is essential for mouse development and is also required for viability in adult animals. Strikingly, even Pold3(+/-) mice were born at sub-Mendelian ratios, and, of those born, some presented hydrocephaly and had a reduced lifespan. In cells, POLD3 deficiency led to replication stress and cell death, which were aggravated by the expression of activated oncogenes. Finally, we show that Pold3 deletion destabilizes all members of the Polδ complex, explaining its major role in DNA replication and the severe impact of its deficiency. Research was funded by Fundacion Botin, Banco Santander, through its Santander Universities Global Division, and by grants from the Spanish Ministry of Economy and Competitiveness (MINECO) (SAF2014-59498-R; SAF2014-57791-REDC), Fundacio La Marato de TV3, the Howard Hughes Medical Institute, and the European Research Council (ERC-617840) to O.F.-C.; by a Marie Curie International Outgoing Fellowshp (IOF) from the FP7 Marie Curie Actions and a grant from MINECO (BFU2014-55168-JIN) that was co-funded by European Regional Development Funds (FEDER) to E.L.; by a grant from MINECO (BFU2013-49153) to J.M.; and by the European Commission (ERC grant ONIDDAC) to T.D.H. Sí
- Published
- 2016
- Full Text
- View/download PDF
44. A Genome-wide CRISPR Screen Identifies CDC25A as a Determinant of Sensitivity to ATR Inhibitors
- Author
-
Vanesa Lafarga, Sagrario Ortega, Sergio Ruiz, Matilde Murga, Maria Vega-Sendino, Oscar Fernandez-Capetillo, Cristina Mayor-Ruiz, Botín Foundation, Ministerio de Economía y Competitividad (España), Fundación La Marató TV3, Howard Hughes Medical Institute, European Research Council, and Fundación La Caixa
- Subjects
0301 basic medicine ,Mitosis ,Antineoplastic Agents ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Transfection ,Article ,Cell Line ,03 medical and health sciences ,RNA interference ,Humans ,cdc25 Phosphatases ,CRISPR ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Molecular Biology ,Embryonic Stem Cells ,Dose-Response Relationship, Drug ,biology ,Nuclear Proteins ,Cell Biology ,Protein-Tyrosine Kinases ,3. Good health ,Cell biology ,Wee1 ,030104 developmental biology ,Drug development ,Drug Resistance, Neoplasm ,Cancer cell ,biology.protein ,RNA Interference ,CRISPR-Cas Systems ,Genome-Wide Association Study ,Signal Transduction - Abstract
One recurring theme in drug development is to exploit synthetic lethal properties as means to preferentially damage the DNA of cancer cells. We and others have previously developed inhibitors of the ATR kinase, shown to be particularly genotoxic for cells expressing certain oncogenes. In contrast, the mechanisms of resistance to ATR inhibitors remain unexplored. We report here on a genome-wide CRISPR-Cas9 screen that identified CDC25A as a major determinant of sensitivity to ATR inhibition. CDC25A-deficient cells resist high doses of ATR inhibitors, which we show is due to their failure to prematurely enter mitosis in response to the drugs. Forcing mitotic entry with WEE1 inhibitors restores the toxicity of ATR inhibitors in CDC25A-deficient cells. With ATR inhibitors now entering the clinic, our work provides a better understanding of the mechanisms by which these compounds kill cells and reveals genetic interactions that could be used for their rational use. We thank the laboratories of Feng Zhang and Kosuke Yusa for sharing all CRISPR-related plasmids used here through Addgene (plasmids 42230, 50946, and 50947) and Edna Fonseca for her comments on the manuscript. Research was funded by Fundacion Botin, Banco Santander, through its Santander Universities Global Division and by grants from the Spanish Ministry of Economy and Competitiveness (MINECO) (SAF2011-23753 and SAF2014-57791-REDC), Fundacio La Marato de TV3, the Howard Hughes Medical Institute, and the European Research Council (ERC-617840) to O.F.-C.; by a PhD fellowship from La Caixa Foundation to C.M.-R.; by grants from MINECO to S.R. (RYC2011-09242 and SAF2013-49147P, this last project co-financed with European FEDER funds); and by a grant from MINECO (SAF2013-44866-R) to S.O. Sí
- Published
- 2016
- Full Text
- View/download PDF
45. USP7 is a SUMO deubiquitinase essential for DNA replication
- Author
-
Oscar Fernandez-Capetillo, Isabel Ruppen, Sara Rodriguez-Acebes, Javier Muñoz, Matilde Murga, Emilio Lecona, Julia Specks, Juan Méndez, and Andrés J. López-Contreras
- Subjects
DNA Replication ,Models, Molecular ,0301 basic medicine ,DNA Repair ,Eukaryotic DNA replication ,Biology ,Pre-replication complex ,Article ,Ubiquitin-Specific Peptidase 7 ,DNA replication factor CDT1 ,03 medical and health sciences ,Replication factor C ,Control of chromosome duplication ,Minichromosome maintenance ,Structural Biology ,Humans ,Molecular Biology ,Genetics ,Ubiquitination ,DNA replication ,Sumoylation ,HCT116 Cells ,Cell biology ,030104 developmental biology ,Small Ubiquitin-Related Modifier Proteins ,biology.protein ,Origin recognition complex ,Ubiquitin-Specific Proteases ,Ubiquitin Thiolesterase ,DNA Damage ,HeLa Cells - Abstract
Post-translational modification of proteins by ubiquitin (Ub) and Ub-like modifiers regulates various aspects of DNA replication. We previously showed that the chromatin around replisomes is rich in SUMO and depleted in Ub, whereas an opposite pattern is observed in mature chromatin. How this SUMO-rich/Ub-low environment is maintained at sites of DNA replication is not known. Here we identify USP7 as a replisome-enriched SUMO deubiquitinase that is essential for DNA replication. By acting on SUMO and SUMOylated proteins, USP7 counteracts their ubiquitination. Chemical inhibition or genetic deletion of USP7 leads to the accumulation of Ub on SUMOylated proteins, which are displaced to chromatin away from replisomes. Our findings provide a model to explain the differential accumulation of SUMO and Ub at replication forks, and identify an essential role of USP7 in DNA replication that should be taken into account for the use of USP7 inhibitors as anticancer agents.
- Published
- 2016
- Full Text
- View/download PDF
46. <scp>TIAR</scp> marks nuclear G2/M transition granules and restricts <scp>CDK</scp> 1 activity under replication stress
- Author
-
Andrés J. López-Contreras, Marius Bruer, Katharina Haneke, Sylvia Erhardt, Lea Roessig, Hsu Min Sung, Georg Stoecklin, Anne Laure Pauleau, Vanesa Lafarga, Sara Rodriguez-Acebes, Juan Méndez, Oscar Fernandez-Capetillo, Oliver J. Gruss, German Cancer Aid, and Deutsche Forschungsgemeinschaft (Alemania)
- Subjects
CDK1 ,G2/M checkpoint ,0303 health sciences ,Cyclin-dependent kinase 1 ,DNA replication ,RNA-binding protein ,Cell cycle ,Biochemistry ,Cell biology ,Chromosome segregation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Prophase ,RNA‐binding protein ,chemistry ,TIAR ,Genetics ,cell cycle ,biological phenomena, cell phenomena, and immunity ,Molecular Biology ,Mitosis ,030217 neurology & neurosurgery ,DNA ,030304 developmental biology - Abstract
The G2/M checkpoint coordinates DNA replication with mitosis and thereby prevents chromosome segregation in the presence of unreplicated or damaged DNA Here, we show that the RNA-binding protein TIAR is essential for the G2/M checkpoint and that TIAR accumulates in nuclear foci in late G2 and prophase in cells suffering from replication stress. These foci, which we named G2/M transition granules (GMGs), occur at low levels in normally cycling cells and are strongly induced by replication stress. In addition to replication stress response proteins, GMGs contain factors involved in RNA metabolism as well as CDK1. Depletion of TIAR accelerates mitotic entry and leads to chromosomal instability in response to replication stress, in a manner that can be alleviated by the concomitant depletion of Cdc25B or inhibition of CDK1. Since TIAR retains CDK1 in GMGs and attenuates CDK1 activity, we propose that the assembly of GMGs may represent a so far unrecognized mechanism that contributes to the activation of the G2/M checkpoint in mammalian cells. We would like to thank Nancy Kedersha and Paul Anderson (both Harvard Medical School, Boston, USA) for sharing plasmids and thoughtful comments on the manuscript, Thomas Hofmann (DKFZ, Heidelberg) for sharing ideas and reagents, Jan Ellenberg (EMBL Heidelberg, Germany) for sharing the HeLa‐H2B/tub cell line, Iain Mattaj (EMBL Heidelberg, Germany) for sharing the anti‐Sm (Y12) antibody, Angus Lamond (University of Dundee, UK) for sharing the anti‐Coilin antibody, Ingrid Hoffmann (DKFZ Heidelberg, Germany) for sharing the mCherry‐CDK1 plasmid, as well as Guillermo de Carcer (CNIO), Ana Losada (CNIO), and Brian Luke (ZMBH, Heidelberg) for thoughtful comments and critical reading of the manuscript. We also thank Holger Lorenz and Diego Megias from the ZMBH and CNIO imaging core facilities for assistance with microscopy and Monika Langloz from the ZMBH FACS facility for help with flow cytometry. We are grateful to Kathrin Bajak for experimental contributions. This work was supported by a Marie‐Curie Intra‐European Fellowship (mirnaAGOddr, grant Nr. 300384) to VL, doctoral fellowships from the Heidelberg Biosciences International Graduate School to H‐MS and MB, grant Nr. 111219 from the Deutsche Krebshilfe to VL and GS, and grant SFB 1036/TP07 from the Deutsche Forschungsgemeinschaft to GS. Sí
- Published
- 2018
- Full Text
- View/download PDF
47. Generalized displacement of DNA- and RNA-binding factors mediates the toxicity of arginine-rich cell-penetrating peptides
- Author
-
Vanesa Lafarga, Irene Díaz-López, Javier Munoz, Iván Ventoso, Jasminka Boskovic, Eduardo Zarzuela, Oscar Fernandez-Capetillo, Misaru Hisaoka, Oleksandra Sirozh, Rafael Fernandez-Leiro, Georg Stoecklin, and Bogdan Jovanovic
- Subjects
0303 health sciences ,Arginine ,biology ,Nucleolus ,Chemistry ,Oligonucleotide ,Cell ,RNA ,010402 general chemistry ,01 natural sciences ,Protamine ,Molecular biology ,0104 chemical sciences ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,medicine.anatomical_structure ,medicine ,biology.protein ,Nucleic acid ,DNA ,030304 developmental biology - Abstract
Due to their capability to transport chemicals or proteins into target cells, cell-penetrating peptides (CPPs) are being developed as therapy delivery tools. However, and despite their interesting properties, arginine-rich CPPs often show toxicity for reasons that remain poorly understood. Using a (PR)n dipeptide repeat that has been linked to amyotrophic-lateral sclerosis (ALS) as a model of an arginine-rich CPP, we here show that the presence of (PR)n leads to a generalized displacement of RNA- and DNA-binding proteins from chromatin and mRNA. Accordingly, any reaction involving nucleic acids such as RNA transcription, translation, splicing and degradation or DNA replication and repair are impaired by the presence of the CPP. Interestingly, the effects of (PR)n are fully mimicked by PROTAMINE, a small arginine-rich protein that displaces histones from chromatin during spermatogenesis. We propose that widespread coating of nucleic acids and consequent displacement of RNA- and DNA-binding factors from chromatin and mRNA accounts for the toxicity of arginine-rich CPPs, including those that have been recently associated to the onset of ALS.
- Published
- 2018
- Full Text
- View/download PDF
48. Targeting ATR in cancer
- Author
-
Oscar Fernandez-Capetillo and Emilio Lecona
- Subjects
0301 basic medicine ,Genome instability ,Indoles ,DNA damage ,General Mathematics ,Morpholines ,Ataxia Telangiectasia Mutated Proteins ,Genomic Instability ,03 medical and health sciences ,Drug Development ,Neoplasms ,Oxazines ,medicine ,Humans ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Sulfonamides ,business.industry ,Kinase ,Applied Mathematics ,Cancer ,Isoxazoles ,medicine.disease ,Clinical trial ,030104 developmental biology ,Pyrimidines ,Drug development ,Pyrazines ,Sulfoxides ,Ataxia-telangiectasia ,Cancer cell ,Cancer research ,Quinolines ,business ,DNA Damage ,Nitroso Compounds - Abstract
The chemical treatment of cancer started with the realization that DNA damaging agents such as mustard gas present notable antitumoural properties. Consequently, early drug development focused on genotoxic chemicals, some of which are still widely used in the clinic. However, the efficacy of such therapies is often limited by the side effects of these drugs on healthy cells. A refinement to this approach is to use compounds that can exploit the presence of DNA damage in cancer cells. Given that replication stress (RS) is a major source of genomic instability in cancer, targeting the RS-response kinase ataxia telangiectasia and Rad3-related protein (ATR) has emerged as a promising alternative. With ATR inhibitors now entering clinical trials, we here revisit the biology behind this strategy and discuss potential biomarkers that could be used for a better selection of patients who respond to therapy.
- Published
- 2018
49. USP7 couples DNA replication termination to mitotic entry
- Author
-
Julia Specks, Vanesa Lafarga, Pablo Valledor, Antonio Galarreta, Oscar Fernandez-Capetillo, Emilio Lecona, and Patricia Ubieto
- Subjects
0303 health sciences ,Cyclin-dependent kinase 1 ,POLD1 ,DNA replication ,Biology ,Cell biology ,Chromosome segregation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,DNA replication termination ,Replisome ,Mitosis ,030217 neurology & neurosurgery ,DNA ,030304 developmental biology - Abstract
SUMMARYTo ensure a faithful segregation of chromosomes, DNA must be fully replicated before mitotic entry. However, how cells sense the completion of DNA replication and to what extent this is linked to the activation of the mitotic machinery remains poorly understood. We previously showed that USP7 is a replisome-associated deubiquitinase with an essential role in DNA replication. Here, we reveal that USP7 inhibition leads to the ubiquitination of MCM7, a hallmark of DNA replication termination. In addition, USP7 inhibition leads to the ubiquitination of additional replisome components such as POLD1, which are displaced from replisomes. Surprisingly, this premature termination of DNA replication occurs concomitant to a generalized activation of CDK1 throughout the entire cell cycle, which impairs chromosome segregation and is toxic for mammalian cells. Accordingly, the toxicity of USP7 inhibitors is alleviated by CDK1 inhibition. Our work sheds light into the mechanism of action of USP7 inhibitors and provides evidence to the concept that DNA replication termination is coupled to the activation of the mitotic program.
- Published
- 2018
- Full Text
- View/download PDF
50. ERF deletion rescues RAS deficiency in mouse embryonic stem cells
- Author
-
Sergio Ruiz, Matthias Drosten, Teresa Olbrich, Sagrario Ortega, Emilio Lecona, Maria Vega-Sendino, Orlando Domínguez, Oscar Fernandez-Capetillo, Cristina Mayor-Ruiz, Mariano Barbacid, Fundación La Caixa, Boehringer Ingelheim Fonds, Botín Foundation, Banco Santander, European Research Council, Ministerio de Economía y Competitividad (España), Unión Europea. Fondo Europeo de Desarrollo Regional (FEDER/ERDF), Fundación La Marató TV3, and Howard Hughes Medical Institute
- Subjects
Pluripotency ,0301 basic medicine ,MAPK/ERK pathway ,2i ,Mice, Nude ,mESCs ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mediator ,Genetics ,medicine ,Animals ,Glycogen synthase ,Enhancer ,Domain family ,Embryonic Stem Cells ,biology ,fungi ,Teratoma ,food and beverages ,Cell Differentiation ,Embryonic stem cell ,Cell biology ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Genes, ras ,ERF ,030220 oncology & carcinogenesis ,biology.protein ,Transcriptional Repressor ,Nucleus ,Gene Deletion ,RAS ,Developmental Biology ,Research Paper - Abstract
MEK inhibition in combination with a glycogen synthase kinase-3β (GSK3β) inhibitor, referred as the 2i condition, favors pluripotency in embryonic stem cells (ESCs). However, the mechanisms by which the 2i condition limits ESC differentiation and whether RAS proteins are involved in this phenomenon remain poorly understood. Here we show that RAS nullyzygosity reduces the growth of mouse ESCs (mESCs) and prohibits their differentiation. Upon RAS deficiency or MEK inhibition, ERF (E twenty-six 2 [Ets2]-repressive factor), a transcriptional repressor from the ETS domain family, translocates to the nucleus, where it binds to the enhancers of pluripotency factors and key RAS targets. Remarkably, deletion of Erf rescues the proliferative defects of RAS-devoid mESCs and restores their capacity to differentiate. Furthermore, we show that Erf loss enables the development of RAS nullyzygous teratomas. In summary, this work reveals an essential role for RAS proteins in pluripotency and identifies ERF as a key mediator of the response to RAS/MEK/ERK inhibition in mESCs. We thank Cian Lynch, Jorge Monsech, and Diego Megias for their help with microarray, ChIP-seq, and high-throughput microscopy analyses. We also thank Dr. Manuel Serrano and Dr. André Nussenzweig for their input on the manuscript, and Dr. Diego Sanz for his support throughout the project. C.M.-R. was funded by a PhD fellowship from La Caixa Foundation, T.O. was funded by a PhD fellowship from the Boehringer Ingelheim Fonds, and S.R. was funded by a Ramon y Cajal contract (RYC-2011-09242). Research was funded by Fundación Botín and Banco Santander through its Santander Universities Global Division; grants from the Spanish Ministry of Economy and Competitiveness (SAF2011-23753 and SAF2014- 57791-REDC; these projects were cofinanced with European Fonds Européen de Développement Économique et Régional [FEDER] funds), Fundació La Marato de TV3, Howard Hughes Medical Institute, and the European Research Council (ERC- 617840) to O.F.-C.; and grants from the Spanish Ministryof Economy and Competitiveness (SAF2013-49147-P and SAF2016-80874-P; these projects were cofinanced with European FEDER funds) to S.R. Author contributions: C.M.-R. and S.R. conducted most of the experiments; T.O. helped with the characterization of RASlox/lox mESCs and with ERF localization studies; E.L. helped with ChIP-seq experiments; M.D., S.O., and M.B. contributed to the work on RAS-deficient cells; M.V.-S. provided technical help; O.D. helped with genomics experiments and bioinformatics analysis; and S.R. and O.F.-C. coordinated the study and wrote the manuscript. Sí
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.