74 results on '"Prekovic, S."'
Search Results
2. The role of TET-mediated DNA hydroxymethylation in prostate cancer
- Author
-
Smeets, E., Lynch, A.G., Prekovic, S., Van den Broeck, T., Moris, L., Helsen, C., Joniau, S., Claessens, F., and Massie, C.E.
- Published
- 2018
- Full Text
- View/download PDF
3. Treatment-induced changes in the androgen receptor axis: Liquid biopsies as diagnostic/prognostic tools for prostate cancer
- Author
-
Prekovic, S., Van den Broeck, T., Moris, L., Smeets, E., Claessens, F., Joniau, S., Helsen, C., and Attard, G.
- Published
- 2018
- Full Text
- View/download PDF
4. Single-cell ATAC and RNA sequencing reveal pre-existing and persistent subpopulations of cells associated with relapse of prostate cancer
- Author
-
Taavitsainen, S, primary, Engedal, N, additional, Cao, S, additional, Handle, F, additional, Erickson, A, additional, Prekovic, S, additional, Wetterskog, D, additional, Tolonen, T, additional, Vuorinen, EM, additional, Kiviaho, A, additional, Nätkin, R, additional, Häkkinen, T, additional, Devlies, W, additional, Henttinen, S, additional, Kaarijärvi, R, additional, Lahnalampi, M, additional, Kaljunen, H, additional, Nowakowska, K, additional, Syvälä, H, additional, Bläuer, M, additional, Cremaschi, P, additional, Claessens, F, additional, Visakorpi, T, additional, Tammela, TLJ, additional, Murtola, T, additional, Granberg, KJ, additional, Lamb, AD, additional, Ketola, K, additional, Mills, IG, additional, Attard, G, additional, Wang, W, additional, Nykter, M, additional, and Urbanucci, A, additional
- Published
- 2021
- Full Text
- View/download PDF
5. Drivers and therapeutic vulnerabilities in AR indifferent anti-androgen resistant prostate cancer cells
- Author
-
Handle, F., primary, Prekovic, S., additional, Helsen, C., additional, Van Den Broeck, T., additional, Smeets, E., additional, Moris, L., additional, Eerlings, R., additional, El Kharraz, S., additional, Urbanucci, A., additional, Mills, I.G., additional, Joniau, S., additional, Attard, G., additional, and Claessens, F., additional
- Published
- 2019
- Full Text
- View/download PDF
6. A CRISPR-Cas9 screen identifies essential CTCF anchor sites for estrogen receptor-driven breast cancer cell proliferation
- Author
-
Korkmaz, Gokhan, Manber, Z, Lopes, R (Rui Filipe Marques), Prekovic, S, Schuurman, K, Kim, Y, Teunissen, H, Flach, K, Wit, E, Galli, GG, Zwart, W, Elkon, R, Agami, Reuven, Korkmaz, Gokhan, Manber, Z, Lopes, R (Rui Filipe Marques), Prekovic, S, Schuurman, K, Kim, Y, Teunissen, H, Flach, K, Wit, E, Galli, GG, Zwart, W, Elkon, R, and Agami, Reuven
- Published
- 2019
7. Molecular underpinnings of enzalutamide resistance
- Author
-
Prekovic, S, primary, Van den Broeck, T, additional, Linder, S, additional, van Royen, M E, additional, Houtsmuller, A B, additional, Handle, F, additional, Joniau, S, additional, Zwart, W, additional, and Claessens, F, additional
- Published
- 2018
- Full Text
- View/download PDF
8. Genomic analysis of primary high-risk prostate cancer tumors with metastatic recurrence identifies antizyme inhibitor 1 as a regulator of cancer cell migration through regulation of collagen expression
- Author
-
Van Den Broeck, T., primary, Moris, L., additional, Gevaert, T., additional, Prekovic, S., additional, Tosco, L., additional, Smeets, E., additional, Lehrer, J., additional, Haddad, Z., additional, Helsen, C., additional, Margrave, J., additional, Boeckx, B., additional, Lambrechts, D., additional, Van Poppel, H., additional, Everaerts, W., additional, Chellisery, J., additional, Erho, N., additional, Buerki, C., additional, Davicioni, E., additional, Joniau, S., additional, and Claessens, F., additional
- Published
- 2018
- Full Text
- View/download PDF
9. A genomic analysis of metastases-prone localized prostate cancer in a European high-risk population
- Author
-
Van Den Broeck, T., primary, Gevaert, T., additional, Prekovic, S., additional, Ong, K., additional, Tosco, L., additional, Moris, L., additional, Smeets, E., additional, Lehrer, J., additional, Haddad, Z., additional, Helsen, C., additional, Margrave, J., additional, Van Poppel, H., additional, Everaerts, W., additional, Erho, N., additional, Buerki, C., additional, Davicioni, E., additional, Joniau, S., additional, and Claessens, F., additional
- Published
- 2017
- Full Text
- View/download PDF
10. Description of the dimerization surface for the ligand-binding domain of the androgen receptor and its role in transcriptional control by agonists and antagonists
- Author
-
Claessens, F., primary, Nadal, M., additional, Prekovic, S., additional, Gallastegui, N., additional, Helsen, C., additional, Abella, M., additional, Zielinska, K., additional, Gay, M., additional, Vilaseca, M., additional, Taules, M., additional, Houtsmuller, A., additional, Van Royen, M., additional, Fuentes-Prior, P., additional, and Estebanez-Perpina, E., additional
- Published
- 2017
- Full Text
- View/download PDF
11. Structure of the homodimeric androgen receptor ligand-binding domain
- Author
-
Nadal, M, Prekovic, S, Gallastegui, N, Helsen, C, Abella, M, Zielinska, K, Gay, M, Vilaseca, M, Taules, M, Houtsmuller, Adriaan, van Royen, Martin, Claessens, F, Fuentes-Prior, P, Estebanez-Perpina, E, Nadal, M, Prekovic, S, Gallastegui, N, Helsen, C, Abella, M, Zielinska, K, Gay, M, Vilaseca, M, Taules, M, Houtsmuller, Adriaan, van Royen, Martin, Claessens, F, Fuentes-Prior, P, and Estebanez-Perpina, E
- Published
- 2017
12. P39 - Drivers and therapeutic vulnerabilities in AR indifferent anti-androgen resistant prostate cancer cells
- Author
-
Handle, F., Prekovic, S., Helsen, C., Van Den Broeck, T., Smeets, E., Moris, L., Eerlings, R., El Kharraz, S., Urbanucci, A., Mills, I.G., Joniau, S., Attard, G., and Claessens, F.
- Published
- 2019
- Full Text
- View/download PDF
13. Multidisciplinary investigation links backward-speech trait and working memory through genetic mutation
- Author
-
Prekovic, S, ?ur?evi?, D, Csifcsák, G, Šveljo, O, Stojkovi?, O, Jankovi?, M, Koprivšek, K, Covill, L, Lu?i?, M, Van der BroeCk, T, Helsen, C, Ceroni, F, Claessens, F, Newbury, D, Prekovic, S, ?ur?evi?, D, Csifcsák, G, Šveljo, O, Stojkovi?, O, Jankovi?, M, Koprivšek, K, Covill, L, Lu?i?, M, Van der BroeCk, T, Helsen, C, Ceroni, F, Claessens, F, and Newbury, D
- Abstract
Case studies of unusual traits can provide unique snapshots of the effects of modified systems. In this study, we report on an individual from a Serbian family with the ability to rapidly, accurately and voluntarily speak backwards. We consider psychological, neural and genetic correlates of this trait to identify specific relevant neural mechanisms and new molecular pathways for working memory and speech-related tasks. EEG data suggest that the effect of word reversal precedes semantic integration of visually presented backward-words, and that event-related potentials above the frontal lobe are affected by both word reversal and the maintenance of backward-words in working memory. fMRI revealed that the left fusiform gyrus may facilitate the production of backward-speech. Exome sequencing identified three novel coding variants of potential significance in the RIC3, RIPK1 and ZBED5 genes. Taken together, our data suggest that, in this individual, the ability to speak backwards is afforded by an extraordinary working memory capacity. We hypothesise that this is served by cholinergic projections from the basal forebrain to the frontal cortex and supported by visual semantic loops within the left fusiform gyrus and that these neural processes may be mediated by a genetic mutation in RIC3; a chaperone for nicotinic acetylcholine receptors.
- Published
- 2016
14. 1073 Prostate cancer copy number score predicts metastatic disease
- Author
-
Van Den Broeck, T., primary, Gevaert, T., additional, Prekovic, S., additional, Smeets, E., additional, Helsen, C., additional, Lambrechts, D., additional, Boeckx, B., additional, Joniau, S., additional, and Claessens, F., additional
- Published
- 2016
- Full Text
- View/download PDF
15. 57 - Genomic analysis of primary high-risk prostate cancer tumors with metastatic recurrence identifies antizyme inhibitor 1 as a regulator of cancer cell migration through regulation of collagen expression
- Author
-
Van Den Broeck, T., Moris, L., Gevaert, T., Prekovic, S., Tosco, L., Smeets, E., Lehrer, J., Haddad, Z., Helsen, C., Margrave, J., Boeckx, B., Lambrechts, D., Van Poppel, H., Everaerts, W., Chellisery, J., Erho, N., Buerki, C., Davicioni, E., Joniau, S., and Claessens, F.
- Published
- 2018
- Full Text
- View/download PDF
16. 483 - A genomic analysis of metastases-prone localized prostate cancer in a European high-risk population
- Author
-
Van Den Broeck, T., Gevaert, T., Prekovic, S., Ong, K., Tosco, L., Moris, L., Smeets, E., Lehrer, J., Haddad, Z., Helsen, C., Margrave, J., Van Poppel, H., Everaerts, W., Erho, N., Buerki, C., Davicioni, E., Joniau, S., and Claessens, F.
- Published
- 2017
- Full Text
- View/download PDF
17. 99 - Description of the dimerization surface for the ligand-binding domain of the androgen receptor and its role in transcriptional control by agonists and antagonists
- Author
-
Claessens, F., Nadal, M., Prekovic, S., Gallastegui, N., Helsen, C., Abella, M., Zielinska, K., Gay, M., Vilaseca, M., Taules, M., Houtsmuller, A., Van Royen, M., Fuentes-Prior, P., and Estebanez-Perpina, E.
- Published
- 2017
- Full Text
- View/download PDF
18. Multidisciplinary investigation links backward-speech trait and working memory through genetic mutation
- Author
-
Prekovic, Stefan, Đurđević, Dušica Filipović, Csifcsák, Gábor, Šveljo, Olivera, Stojković, Oliver, Janković, Milica, Koprivšek, Katarina, Covill, Laura E, Lučić, Milos, Van den Broeck, Thomas, Helsen, Christine, Ceroni, Fabiola, Claessens, Frank, Newbury, Dianne F, Prekovic S., Durdevic D.F., Csifcsak G., Sveljo O., Stojkovic O., Jankovic M., Koprivsek K., Covill L.E., Lucic M., Van Den Broeck T., Helsen C., Ceroni F., Claessens F., and Newbury D.F.
- Subjects
Adult ,Male ,Quantitative Trait Loci ,Intracellular Signaling Peptides and Proteins ,Middle Aged ,Article ,Semantics ,Memory, Short-Term ,Reading ,Receptor-Interacting Protein Serine-Threonine Kinases ,Mutation ,Humans ,Female ,Serbia ,Backward-speech, Language, Working memory, RIC3 - Abstract
Case studies of unusual traits can provide unique snapshots of the effects of modified systems. In this study, we report on an individual from a Serbian family with the ability to rapidly, accurately and voluntarily speak backwards. We consider psychological, neural and genetic correlates of this trait to identify specific relevant neural mechanisms and new molecular pathways for working memory and speech-related tasks. EEG data suggest that the effect of word reversal precedes semantic integration of visually presented backward-words, and that event-related potentials above the frontal lobe are affected by both word reversal and the maintenance of backward-words in working memory. fMRI revealed that the left fusiform gyrus may facilitate the production of backward-speech. Exome sequencing identified three novel coding variants of potential significance in the RIC3, RIPK1 and ZBED5 genes. Taken together, our data suggest that, in this individual, the ability to speak backwards is afforded by an extraordinary working memory capacity. We hypothesise that this is served by cholinergic projections from the basal forebrain to the frontal cortex and supported by visual semantic loops within the left fusiform gyrus and that these neural processes may be mediated by a genetic mutation in RIC3; a chaperone for nicotinic acetylcholine receptors. ispartof: Scientific Reports vol:6 issue:1 ispartof: location:England status: Published online
- Published
- 2016
- Full Text
- View/download PDF
19. Emerging roles of cohesin-STAG2 in cancer.
- Author
-
Scott JS, Al Ayadi L, Epeslidou E, van Scheppingen RH, Mukha A, Kaaij LJT, Lutz C, and Prekovic S
- Subjects
- Humans, Animals, DNA Repair genetics, Gene Expression Regulation, Neoplastic, Chromatin metabolism, Chromatin genetics, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Cohesins, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology
- Abstract
Cohesin, a crucial regulator of genome organisation, plays a fundamental role in maintaining chromatin architecture as well as gene expression. Among its subunits, STAG2 stands out because of its frequent deleterious mutations in various cancer types, such as bladder cancer and melanoma. Loss of STAG2 function leads to significant alterations in chromatin structure, disrupts transcriptional regulation, and impairs DNA repair pathways. In this review, we explore the molecular mechanisms underlying cohesin-STAG2 function, highlighting its roles in healthy cells and its contributions to cancer biology, showing how STAG2 dysfunction promotes tumourigenesis and presents opportunities for targeted therapeutic interventions., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2025
- Full Text
- View/download PDF
20. NAC regulates metabolism and cell fate in intestinal stem cells.
- Author
-
Ramalho S, Alkan F, Prekovic S, Jastrzebski K, Barberà EP, Hoekman L, Altelaar M, de Heus C, Liv N, Rodríguez-Colman MJ, Yilmaz M, van der Kammen R, Fedry J, de Gooijer MC, Suijkerbuijk SJE, Faller WJ, and Silva J
- Subjects
- Animals, Mice, Cell Differentiation, Organoids metabolism, Organoids cytology, Oxidative Phosphorylation, Intestinal Mucosa metabolism, Intestinal Mucosa cytology, Mitochondrial Proteins metabolism, Mitochondrial Proteins genetics, Stem Cells metabolism, Stem Cells cytology, Mitochondria metabolism, Ribosomes metabolism, Intestines cytology
- Abstract
Intestinal stem cells (ISCs) face the challenge of integrating metabolic demands with unique regenerative functions. Studies have shown an intricate interplay between metabolism and stem cell capacity; however, it is still not understood how this process is regulated. Combining ribosome profiling and CRISPR screening in intestinal organoids, we identify the nascent polypeptide-associated complex (NAC) as a key mediator of this process. Our findings suggest that NAC is responsible for relocalizing ribosomes to the mitochondria and regulating ISC metabolism. Upon NAC inhibition, intestinal cells show decreased import of mitochondrial proteins, which are needed for oxidative phosphorylation, and, consequently, enable the cell to maintain a stem cell identity. Furthermore, we show that overexpression of NACα is sufficient to drive mitochondrial respiration and promote ISC identity. Ultimately, our results reveal the pivotal role of NAC in regulating ribosome localization, mitochondrial metabolism, and ISC function, providing insights into the potential mechanism behind it.
- Published
- 2025
- Full Text
- View/download PDF
21. The complex landscape of luminal breast cancer.
- Author
-
Lutz C, Messal HA, Vareslija D, and Prekovic S
- Subjects
- Humans, Female, Animals, Breast Neoplasms pathology, Breast Neoplasms metabolism, Receptors, Estrogen metabolism
- Abstract
The breast epithelium, vital for mammary gland function, is influenced by oestrogen through the oestrogen receptor (ER) signalling pathway. Luminal breast cancer (BC), characterised by ER expression, comprises the majority of all BCs and presents significant clinical challenges due to therapy resistance and recurrence. Despite advancements in understanding luminal disease, improving long-term survival and reducing relapse of BC patients by predicting therapy efficacy and understanding resistance mechanisms remain critical challenges. This review discusses luminal BC biology, focusing on the molecular classification of primary disease, metastatic spread, and experimental models.
- Published
- 2024
- Full Text
- View/download PDF
22. IFNγ induces epithelial reprogramming driving CXCL11-mediated T cell migration.
- Author
-
Cutilli A, Jansen SA, Paolucci F, van Hoesel M, Fredericks CL, Mulder TAM, Chalkiadakis T, Mokry M, Prekovic S, Mocholi E, Lindemans CA, and Coffer PJ
- Abstract
The cytokine interferon-gamma (IFNγ) plays a multifaceted role in intestinal immune responses ranging from anti- to pro-inflammatory depending on the setting. Here, using a 3D co-culture system based on human intestinal epithelial organoids, we explore the capacity of IFNγ-exposure to reprogram intestinal epithelia and thereby directly modulate lymphocyte responses. IFNγ treatment of organoids led to transcriptional reprogramming, marked by a switch to a pro-inflammatory gene expression profile, including transcriptional upregulation of the chemokines CXCL9, CXCL10, and CXCL11. Proteomic analysis of organoid-conditioned medium post-treatment confirmed chemokine secretion. IFNγ-treatment of organoids led to enhanced T cell migration in a CXCL11-dependent manner without affecting T cell activation status. Taken together, our results suggest a specific role for CXCL11 in T cell recruitment that could be targeted to prevent T cell trafficking to the inflamed intestine., (© The Author(s) 2024. Published by Oxford University Press on behalf of Society for Leukocyte Biology.)
- Published
- 2024
- Full Text
- View/download PDF
23. Mechanisms that clear mutations drive field cancerization in mammary tissue.
- Author
-
Ciwinska M, Messal HA, Hristova HR, Lutz C, Bornes L, Chalkiadakis T, Harkes R, Langedijk NSM, Hutten SJ, Menezes RX, Jonkers J, Prekovic S, Simons BD, Scheele CLGJ, and van Rheenen J
- Subjects
- Animals, Female, Mice, BRCA1 Protein deficiency, BRCA1 Protein genetics, BRCA1 Protein metabolism, Cell Lineage genetics, Cell Self Renewal genetics, Clone Cells cytology, Clone Cells metabolism, Clone Cells pathology, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Estrous Cycle, Stem Cells cytology, Stem Cells metabolism, Stem Cells pathology, Cell Transformation, Neoplastic genetics, Mammary Glands, Animal cytology, Mammary Glands, Animal pathology, Mammary Glands, Animal metabolism, Mutation
- Abstract
Oncogenic mutations are abundant in the tissues of healthy individuals, but rarely form tumours
1-3 . Yet, the underlying protection mechanisms are largely unknown. To resolve these mechanisms in mouse mammary tissue, we use lineage tracing to map the fate of wild-type and Brca1-/- ;Trp53-/- cells, and find that both follow a similar pattern of loss and spread within ducts. Clonal analysis reveals that ducts consist of small repetitive units of self-renewing cells that give rise to short-lived descendants. This offers a first layer of protection as any descendants, including oncogenic mutant cells, are constantly lost, thereby limiting the spread of mutations to a single stem cell-descendant unit. Local tissue remodelling during consecutive oestrous cycles leads to the cooperative and stochastic loss and replacement of self-renewing cells. This process provides a second layer of protection, leading to the elimination of most mutant clones while enabling the minority that by chance survive to expand beyond the stem cell-descendant unit. This leads to fields of mutant cells spanning large parts of the epithelial network, predisposing it for transformation. Eventually, clone expansion becomes restrained by the geometry of the ducts, providing a third layer of protection. Together, these mechanisms act to eliminate most cells that acquire somatic mutations at the expense of driving the accelerated expansion of a minority of cells, which can colonize large areas, leading to field cancerization., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
24. Differential transcriptional invasion signatures from patient derived organoid models define a functional prognostic tool for head and neck cancer.
- Author
-
Haughton PD, Haakma W, Chalkiadakis T, Breimer GE, Driehuis E, Clevers H, Willems S, Prekovic S, and Derksen PWB
- Subjects
- Humans, Prognosis, Gene Expression Regulation, Neoplastic, Cell Movement genetics, Cell Line, Tumor, Transcription Factors genetics, Transcription Factors metabolism, YAP-Signaling Proteins, Organoids pathology, Organoids metabolism, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Neoplasm Invasiveness, Squamous Cell Carcinoma of Head and Neck genetics, Squamous Cell Carcinoma of Head and Neck pathology
- Abstract
Clinical outcome for patients suffering from HPV-negative head and neck squamous cell carcinoma (HNSCC) remains poor. This is mostly due to highly invasive tumors that cause loco-regional relapses after initial therapeutic intervention and metastatic outgrowth. The molecular pathways governing the detrimental invasive growth modes in HNSCC remain however understudied. Here, we have established HNSCC patient derived organoid (PDO) models that recapitulate 3-dimensional invasion in vitro. Single cell mRNA sequencing was applied to study the differences between non-invasive and invasive conditions, and in a collective versus single cell invading PDO model. Differential expression analysis under invasive conditions in Collagen gels reveals an overall upregulation of a YAP-centered transcriptional program, irrespective of the invasion mode. However, we find that collectively invading HNSCC PDO cells show elevated levels of YAP transcription targets when compared to single cell invasion. Also, collectively invading cells are characterized by increased nuclear translocation of YAP within the invasive strands, which coincides with Collagen-I matrix alignment at the invasive front. Using gene set enrichment analysis, we identify immune cell-like migratory pathways in the single cell invading HNSCC PDO, while collective invasion is characterized by overt upregulation of adhesion and migratory pathways. Lastly, based on clinical head and neck cancer cohorts, we demonstrate that the identified collective invasion signature provides a candidate prognostic platform for survival in HNSCC. By uncoupling collective and single cell invasive programs, we have established invasion signatures that may guide new therapeutic options., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
25. Unraveling the molecular interactions between α7 nicotinic receptor and a RIC3 variant associated with backward speech.
- Author
-
Pradhan A, Mounford H, Peixinho J, Rea E, Epeslidou E, Scott JS, Cull J, Maxwell S, Webster R, Beeson D, Dong YY, Prekovic S, Bermudez I, and Newbury DF
- Subjects
- Humans, Acetylcholinesterase metabolism, alpha7 Nicotinic Acetylcholine Receptor metabolism, Cell Membrane metabolism, Intracellular Signaling Peptides and Proteins metabolism, Speech, Receptors, Nicotinic genetics
- Abstract
Recent work putatively linked a rare genetic variant of the chaperone Resistant to Inhibitors of acetylcholinesterase (RIC3) (NM_024557.4:c.262G > A, NP_078833.3:p.G88R) to a unique ability to speak backwards, a language skill that is associated with exceptional working memory capacity. RIC3 is important for the folding, maturation, and functional expression of α7 nicotinic acetylcholine receptors (nAChR). We compared and contrasted the effects of RIC3G88R on assembly, cell surface expression, and function of human α7 receptors using fluorescent protein tagged α7 nAChR and Förster resonance energy transfer (FRET) microscopy imaging in combination with functional assays and
125 I-α-bungarotoxin binding. As expected, the wild-type RIC3 protein was found to increase both cell surface and functional expression of α7 receptors. In contrast, the variant form of RIC3 decreased both. FRET analysis showed that RICG88R increased the interactions between RIC3 and α7 protein in the endoplasmic reticulum. These results provide interesting and novel data to show that a RIC3 variant alters the interaction of RIC3 and α7, which translates to decreased cell surface and functional expression of α7 nAChR., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
26. Proteomics on malignant pleural effusions reveals ERα loss in metastatic breast cancer associates with SGK1-NDRG1 deregulation.
- Author
-
Mayayo-Peralta I, Debets DO, Prekovic S, Schuurman K, Beerthuijzen S, Almekinders M, Sanders J, Moelans CB, Saleiro S, Wesseling J, van Diest PJ, Henrique R, Jerónimo C, Altelaar M, and Zwart W
- Subjects
- Female, Humans, Estrogen Receptor alpha metabolism, Glucocorticoids therapeutic use, Proteomics, Breast Neoplasms pathology, Pleural Effusion, Malignant
- Abstract
Breast cancer (BCa) is a highly heterogeneous disease, with hormone receptor status being a key factor in patient prognostication and treatment decision-making. The majority of primary tumours are positive for oestrogen receptor alpha (ERα), which plays a key role in tumorigenesis and disease progression, and represents the major target for treatment of BCa. However, around one-third of patients with ERα-positive BCa relapse and progress into the metastatic stage, with 20% of metastatic cases characterised by loss of ERα expression after endocrine treatment, known as ERα-conversion. It remains unclear whether ERα-converted cancers are biologically similar to bona fide ERα-negative disease and which signalling cascades compensate for ERα loss and drive tumour progression. To better understand the biological changes that occur in metastatic BCa upon ERα loss, we performed (phospho)proteomics analysis of 47 malignant pleural effusions derived from 37 BCa patients, comparing ERα-positive, ERα-converted and ERα-negative cases. Our data revealed that the loss of ERα-dependency in this metastatic site leads to only a partial switch to an ERα-negative molecular phenotype, with preservation of a luminal-like proteomic landscape. Furthermore, we found evidence for decreased activity of several key kinases, including serum/glucocorticoid regulated kinase 1 (SGK1), in ERα-converted metastases. Loss of SGK1 substrate phosphorylation may compensate for the loss of ERα-dependency in advanced disease and exposes a potential therapeutic vulnerability that may be exploited in treating these patients., (© 2023 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
27. Luminal breast cancer identity is determined by loss of glucocorticoid receptor activity.
- Author
-
Prekovic S, Chalkiadakis T, Roest M, Roden D, Lutz C, Schuurman K, Opdam M, Hoekman L, Abbott N, Tesselaar T, Wajahat M, Dwyer AR, Mayayo-Peralta I, Gomez G, Altelaar M, Beijersbergen R, Győrffy B, Young L, Linn S, Jonkers J, Tilley W, Hickey T, Vareslija D, Swarbrick A, and Zwart W
- Subjects
- Female, Humans, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Breast Neoplasms genetics, Breast Neoplasms drug therapy
- Abstract
Glucocorticoid receptor (GR) is a transcription factor that plays a crucial role in cancer biology. In this study, we utilized an in silico-designed GR activity signature to demonstrate that GR relates to the proliferative capacity of numerous primary cancer types. In breast cancer, the GR activity status determines luminal subtype identity and has implications for patient outcomes. We reveal that GR engages with estrogen receptor (ER), leading to redistribution of ER on the chromatin. Notably, GR activation leads to upregulation of the ZBTB16 gene, encoding for a transcriptional repressor, which controls growth in ER-positive breast cancer and associates with prognosis in luminal A patients. In relation to ZBTB16's repressive nature, GR activation leads to epigenetic remodeling and loss of histone acetylation at sites proximal to cancer-driving genes. Based on these findings, epigenetic inhibitors reduce viability of ER-positive breast cancer cells that display absence of GR activity. Our findings provide insights into how GR controls ER-positive breast cancer growth and may have implications for patients' prognostication and provide novel therapeutic candidates for breast cancer treatment., (© 2023 Netherlands Cancer Institute. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
28. The 2023 generation.
- Author
-
Achinger-Kawecka J, Correa S, Hu J, Li G, Lindeboom RGH, Misale S, Monkkonen T, Nirmal AJ, Prekovic S, Sinha S, Trigos AS, and Watson CJ
- Published
- 2023
- Full Text
- View/download PDF
29. Perturbations in 3D genome organization can promote acquired drug resistance.
- Author
-
Manjón AG, Manzo SG, Prekovic S, Potgeter L, van Schaik T, Liu NQ, Flach K, Peric-Hupkes D, Joosten S, Teunissen H, Friskes A, Ilic M, Hintzen D, Franceschini-Santos VH, Zwart W, de Wit E, van Steensel B, and Medema RH
- Subjects
- Humans, Paclitaxel pharmacology, Drug Resistance, Multiple genetics, DNA Methylation genetics, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Neoplasms genetics
- Abstract
Acquired drug resistance is a major problem in the treatment of cancer. hTERT-immortalized, untransformed RPE-1 cells can acquire resistance to Taxol by derepressing the ABCB1 gene, encoding for the multidrug transporter P-gP. Here, we investigate how the ABCB1 gene is derepressed. ABCB1 activation is associated with reduced H3K9 trimethylation, increased H3K27 acetylation, and ABCB1 displacement from the nuclear lamina. While altering DNA methylation and H3K27 methylation had no major impact on ABCB1 expression, nor did it promote resistance, disrupting the nuclear lamina component Lamin B Receptor did promote the acquisition of a Taxol-resistant phenotype in a subset of cells. CRISPRa-mediated gene activation supported the notion that lamina dissociation influences ABCB1 derepression. We propose a model in which nuclear lamina dissociation of a repressed gene allows for its activation, implying that deregulation of the 3D genome topology could play an important role in tumor evolution and the acquisition of drug resistance., Competing Interests: Declaration of interests E.d.W. is a co-founder of Cergentis B.V., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. Inhibiting the Glucocorticoid Receptor to Enhance Chemotherapy Response.
- Author
-
Prekovic S and Zwart W
- Subjects
- Humans, Signal Transduction, Glucocorticoids, Receptors, Glucocorticoid, Dexamethasone pharmacology
- Published
- 2023
- Full Text
- View/download PDF
31. PAXIP1 and STAG2 converge to maintain 3D genome architecture and facilitate promoter/enhancer contacts to enable stress hormone-dependent transcription.
- Author
-
Mayayo-Peralta I, Gregoricchio S, Schuurman K, Yavuz S, Zaalberg A, Kojic A, Abbott N, Geverts B, Beerthuijzen S, Siefert J, Severson TM, van Baalen M, Hoekman L, Lieftink C, Altelaar M, Beijersbergen RL, Houtsmuller AB, Prekovic S, and Zwart W
- Abstract
How steroid hormone receptors (SHRs) regulate transcriptional activity remains partly understood. Upon activation, SHRs bind the genome together with a co-regulator repertoire, crucial to induce gene expression. However, it remains unknown which components of the SHR-recruited co-regulator complex are essential to drive transcription following hormonal stimuli. Through a FACS-based genome-wide CRISPR screen, we functionally dissected the Glucocorticoid Receptor (GR) complex. We describe a functional cross-talk between PAXIP1 and the cohesin subunit STAG2, critical for regulation of gene expression by GR. Without altering the GR cistrome, PAXIP1 and STAG2 depletion alter the GR transcriptome, by impairing the recruitment of 3D-genome organization proteins to the GR complex. Importantly, we demonstrate that PAXIP1 is required for stability of cohesin on chromatin, its localization to GR-occupied sites, and maintenance of enhancer-promoter interactions. In lung cancer, where GR acts as tumor suppressor, PAXIP1/STAG2 loss enhances GR-mediated tumor suppressor activity by modifying local chromatin interactions. All together, we introduce PAXIP1 and STAG2 as novel co-regulators of GR, required to maintain 3D-genome architecture and drive the GR transcriptional programme following hormonal stimuli., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
32. Crosstalk between glucocorticoid and mineralocorticoid receptors boosts glucocorticoid-induced killing of multiple myeloma cells.
- Author
-
Clarisse D, Prekovic S, Vlummens P, Staessens E, Van Wesemael K, Thommis J, Fijalkowska D, Acke G, Zwart W, Beck IM, Offner F, and De Bosscher K
- Subjects
- Humans, Receptors, Mineralocorticoid genetics, Dexamethasone pharmacology, Dexamethasone metabolism, Dexamethasone therapeutic use, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Spironolactone therapeutic use, Glucocorticoids pharmacology, Multiple Myeloma drug therapy, Multiple Myeloma genetics
- Abstract
The glucocorticoid receptor (GR) is a crucial drug target in multiple myeloma as its activation with glucocorticoids effectively triggers myeloma cell death. However, as high-dose glucocorticoids are also associated with deleterious side effects, novel approaches are urgently needed to improve GR action in myeloma. Here, we reveal a functional crosstalk between GR and the mineralocorticoid receptor (MR) that plays a role in improved myeloma cell killing. We show that the GR agonist dexamethasone (Dex) downregulates MR levels in a GR-dependent way in myeloma cells. Co-treatment of Dex with the MR antagonist spironolactone (Spi) enhances Dex-induced cell killing in primary, newly diagnosed GC-sensitive myeloma cells. In a relapsed GC-resistant setting, Spi alone induces distinct myeloma cell killing. On a mechanistic level, we find that a GR-MR crosstalk likely arises from an endogenous interaction between GR and MR in myeloma cells. Quantitative dimerization assays show that Spi reduces Dex-induced GR-MR heterodimerization and completely abolishes Dex-induced MR-MR homodimerization, while leaving GR-GR homodimerization intact. Unbiased transcriptomics analyses reveal that c-myc and many of its target genes are downregulated most by combined Dex-Spi treatment. Proteomics analyses further identify that several metabolic hallmarks are modulated most by this combination treatment. Finally, we identified a subset of Dex-Spi downregulated genes and proteins that may predict prognosis in the CoMMpass myeloma patient cohort. Our study demonstrates that GR-MR crosstalk is therapeutically relevant in myeloma as it provides novel strategies for glucocorticoid-based dose-reduction., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
33. A genome-wide CRISPR screen in human prostate cancer cells reveals drivers of macrophage-mediated cell killing and positions AR as a tumor-intrinsic immunomodulator.
- Author
-
Zaalberg A, Minnee E, Mayayo-Peralta I, Schuurman K, Gregoricchio S, van Schaik TA, Hoekman L, Li D, Corey E, Janssen H, Lieftink C, Prekovic S, Altelaar M, Nelson PS, Beijersbergen RL, Zwart W, and Bergman A
- Abstract
The crosstalk between prostate cancer (PCa) cells and the tumor microenvironment plays a pivotal role in disease progression and metastasis and could provide novel opportunities for patient treatment. Macrophages are the most abundant immune cells in the prostate tumor microenvironment (TME) and are capable of killing tumor cells. To identify genes in the tumor cells that are critical for macrophage-mediated killing, we performed a genome-wide co-culture CRISPR screen and identified AR, PRKCD, and multiple components of the NF-κB pathway as hits, whose expression in the tumor cell are essential for being targeted and killed by macrophages. These data position AR signaling as an immunomodulator, and confirmed by androgen-deprivation experiments, that rendered hormone-deprived tumor cells resistant to macrophage-mediated killing. Proteomic analyses showed a downregulation of oxidative phosphorylation in the PRKCD- and IKBKG-KO cells compared to the control, suggesting impaired mitochondrial function, which was confirmed by electron microscopy analyses. Furthermore, phosphoproteomic analyses revealed that all hits impaired ferroptosis signaling, which was validated transcriptionally using samples from a neoadjuvant clinical trial with the AR-inhibitor enzalutamide. Collectively, our data demonstrate that AR functions together with the PRKCD and the NF-κB pathway to evade macrophage-mediated killing. As hormonal intervention represents the mainstay therapy for treatment of prostate cancer patients, our findings may have direct implications and provide a plausible explanation for the clinically observed persistence of tumor cells despite androgen deprivation therapy.
- Published
- 2023
- Full Text
- View/download PDF
34. The Tumor Coagulome as a Transcriptional Target and a Potential Effector of Glucocorticoids in Human Cancers.
- Author
-
Racine F, Louandre C, Godin C, Chatelain B, Prekovic S, Zwart W, Galmiche A, and Saidak Z
- Abstract
Background: The coagulome, defined as the repertoire of genes that locally regulate coagulation and fibrinolysis, is a key determinant of vascular thromboembolic complications of cancer. In addition to vascular complications, the coagulome may also regulate the tumor microenvironment (TME). Glucocorticoids are key hormones that mediate cellular responses to various stresses and exert anti-inflammatory effects. We addressed the effects of glucocorticoids on the coagulome of human tumors by investigating interactions with Oral Squamous Cell Carcinoma, Lung Adenocarcinoma, and Pancreatic Adenocarcinoma tumor types., Methods: We analyzed the regulation of three essential coagulome components, i.e., the tissue factor (TF), urokinase-type plasminogen activator (uPA), and plasminogen activator inhibitor-1 (PAI-1) in cancer cell lines exposed to specific agonists of the glucocorticoid receptor (GR) (dexamethasone and hydrocortisone). We used QPCR, immunoblots, small-interfering RNA, Chromatin immunoprecipitation sequencing (ChIPseq) and genomic data from whole tumor and single-cell analyses., Results: Glucocorticoids modulate the coagulome of cancer cells through a combination of indirect and direct transcriptional effects. Dexamethasone directly increased PAI-1 expression in a GR-dependent manner. We confirmed the relevance of these findings in human tumors, where high GR activity/high SERPINE1 expression corresponded to a TME enriched in active fibroblasts and with a high TGF-β response., Conclusion: The transcriptional regulation of the coagulome by glucocorticoids that we report may have vascular consequences and account for some of the effects of glucocorticoids on the TME.
- Published
- 2023
- Full Text
- View/download PDF
35. Enhancer profiling identifies epigenetic markers of endocrine resistance and reveals therapeutic options for metastatic castration-resistant prostate cancer patients.
- Author
-
Severson TM, Zhu Y, Prekovic S, Schuurman K, Nguyen HM, Brown LG, Hakkola S, Kim Y, Kneppers J, Linder S, Stelloo S, Lieftink C, van der Heijden M, Nykter M, van der Noort V, Sanders J, Morris B, Jenster G, van Leenders GJ, Pomerantz M, Freedman ML, Beijersbergen RL, Urbanucci A, Wessels L, Corey E, Zwart W, and Bergman AM
- Abstract
Androgen Receptor (AR) signaling inhibitors, including enzalutamide, are treatment options for patients with metastatic castration-resistant prostate cancer (mCRPC), but resistance inevitably develops. Using metastatic samples from a prospective phase II clinical trial, we epigenetically profiled enhancer/promoter activities with H3K27ac chromatin immunoprecipitation followed by sequencing, before and after AR-targeted therapy. We identified a distinct subset of H3K27ac-differentially marked regions that associated with treatment responsiveness. These data were successfully validated in mCRPC patient-derived xenograft models (PDX). In silico analyses revealed HDAC3 as a critical factor that can drive resistance to hormonal interventions, which we validated in vitro . Using cell lines and mCRPC PDX tumors in vitro , we identified drug-drug synergy between enzalutamide and the pan-HDAC inhibitor vorinostat, providing therapeutic proof-of-concept. These findings demonstrate rationale for new therapeutic strategies using a combination of AR and HDAC inhibitors to improve patient outcome in advanced stages of mCRPC.
- Published
- 2023
- Full Text
- View/download PDF
36. Mammalian life depends on two distinct pathways of DNA damage tolerance.
- Author
-
Buoninfante OA, Pilzecker B, Spanjaard A, de Groot D, Prekovic S, Song JY, Lieftink C, Ayidah M, Pritchard CEJ, Vivié J, Mcgrath KE, Huijbers IJ, Philipsen S, von Lindern M, Zwart W, Beijersbergen RL, Palis J, van den Berk PCM, and Jacobs H
- Subjects
- Animals, DNA Repair, DNA Replication, Hematopoietic Stem Cells metabolism, Mammals metabolism, Proliferating Cell Nuclear Antigen metabolism, Ubiquitination, DNA Damage
- Abstract
DNA damage threatens genomic integrity and instigates stem cell failure. To bypass genotoxic lesions during replication, cells employ DNA damage tolerance (DDT), which is regulated via PCNA ubiquitination and REV1. DDT is conserved in all domains of life, yet its relevance in mammals remains unclear. Here, we show that inactivation of both PCNA-ubiquitination and REV1 results in embryonic and adult lethality, and the accumulation of DNA damage in hematopoietic stem and progenitor cells (HSPCs) that ultimately resulted in their depletion. Our results reveal the crucial relevance of DDT in the maintenance of stem cell compartments and mammalian life in unperturbed conditions.
- Published
- 2023
- Full Text
- View/download PDF
37. Ribosome impairment regulates intestinal stem cell identity via ZAKɑ activation.
- Author
-
Silva J, Alkan F, Ramalho S, Snieckute G, Prekovic S, Garcia AK, Hernández-Pérez S, van der Kammen R, Barnum D, Hoekman L, Altelaar M, Zwart W, Suijkerbuijk SJE, Bekker-Jensen S, and Faller WJ
- Subjects
- Animals, Intestine, Small metabolism, Intestines, Mice, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Ribosomes metabolism, Intestinal Mucosa metabolism, Stem Cells metabolism
- Abstract
The small intestine is a rapidly proliferating organ that is maintained by a small population of Lgr5-expressing intestinal stem cells (ISCs). However, several Lgr5-negative ISC populations have been identified, and this remarkable plasticity allows the intestine to rapidly respond to both the local environment and to damage. However, the mediators of such plasticity are still largely unknown. Using intestinal organoids and mouse models, we show that upon ribosome impairment (driven by Rptor deletion, amino acid starvation, or low dose cyclohexamide treatment) ISCs gain an Lgr5-negative, fetal-like identity. This is accompanied by a rewiring of metabolism. Our findings suggest that the ribosome can act as a sensor of nutrient availability, allowing ISCs to respond to the local nutrient environment. Mechanistically, we show that this phenotype requires the activation of ZAKɑ, which in turn activates YAP, via SRC. Together, our data reveals a central role for ribosome dynamics in intestinal stem cells, and identify the activation of ZAKɑ as a critical mediator of stem cell identity., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
38. Ribociclib Induces Broad Chemotherapy Resistance and EGFR Dependency in ESR1 Wildtype and Mutant Breast Cancer.
- Author
-
Mayayo-Peralta I, Faggion B, Hoekman L, Morris B, Lieftink C, Goldsbrough I, Buluwela L, Siefert JC, Post H, Altelaar M, Beijersbergen R, Ali S, Zwart W, and Prekovic S
- Abstract
While endocrine therapy is highly effective for the treatment of oestrogen receptor-α (ERα)-positive breast cancer, a significant number of patients will eventually experience disease progression and develop treatment-resistant, metastatic cancer. The majority of resistant tumours remain dependent on ERα-action, with activating ESR1 gene mutations occurring in 15-40% of advanced cancers. Therefore, there is an urgent need to discover novel effective therapies that can eradicate cancer cells with aberrant ERα and to understand the cellular response underlying their action. Here, we evaluate the response of MCF7-derived, CRISPR-Cas9-generated cell lines expressing mutant ERα (Y537S) to a large number of drugs. We report sensitivity to numerous clinically approved inhibitors, including CDK4/6 inhibitor ribociclib, which is a standard-of-care therapy in the treatment of metastatic ERα-positive breast cancer and currently under evaluation in the neoadjuvant setting. Ribociclib treatment induces senescence in both wildtype and mutant ERα breast cancer models and leads to a broad-range drug tolerance. Strikingly, viability of cells undergoing ribociclib-induced cellular senescence is maintained via engagement of EGFR signalling, which may be therapeutically exploited in both wildtype and mutant ERα-positive breast cancer. Our study highlights a wide-spread reduction in sensitivity to anti-cancer drugs accompanied with an acquired vulnerability to EGFR inhibitors following CDK4/6 inhibitor treatment.
- Published
- 2021
- Full Text
- View/download PDF
39. The androgen receptor depends on ligand-binding domain dimerization for transcriptional activation.
- Author
-
El Kharraz S, Dubois V, van Royen ME, Houtsmuller AB, Pavlova E, Atanassova N, Nguyen T, Voet A, Eerlings R, Handle F, Prekovic S, Smeets E, Moris L, Devlies W, Ohlsson C, Poutanen M, Verstrepen KJ, Carmeliet G, Launonen KM, Helminen L, Palvimo JJ, Libert C, Vanderschueren D, Helsen C, and Claessens F
- Subjects
- Animals, Binding Sites genetics, Dimerization, Ligands, Male, Mice, Transcriptional Activation, Receptors, Androgen chemistry, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
Whereas dimerization of the DNA-binding domain of the androgen receptor (AR) plays an evident role in recognizing bipartite response elements, the contribution of the dimerization of the ligand-binding domain (LBD) to the correct functioning of the AR remains unclear. Here, we describe a mouse model with disrupted dimerization of the AR LBD (AR
Lmon/Y ). The disruptive effect of the mutation is demonstrated by the feminized phenotype, absence of male accessory sex glands, and strongly affected spermatogenesis, despite high circulating levels of testosterone. Testosterone replacement studies in orchidectomized mice demonstrate that androgen-regulated transcriptomes in ARLmon/Y mice are completely lost. The mutated AR still translocates to the nucleus and binds chromatin, but does not bind to specific AR binding sites. In vitro studies reveal that the mutation in the LBD dimer interface also affects other AR functions such as DNA binding, ligand binding, and co-regulator binding. In conclusion, LBD dimerization is crucial for the development of AR-dependent tissues through its role in transcriptional regulation in vivo. Our findings identify AR LBD dimerization as a possible target for AR inhibition., (© 2021 The Authors.)- Published
- 2021
- Full Text
- View/download PDF
40. Glucocorticoid receptor triggers a reversible drug-tolerant dormancy state with acquired therapeutic vulnerabilities in lung cancer.
- Author
-
Prekovic S, Schuurman K, Mayayo-Peralta I, Manjón AG, Buijs M, Yavuz S, Wellenstein MD, Barrera A, Monkhorst K, Huber A, Morris B, Lieftink C, Chalkiadakis T, Alkan F, Silva J, Győrffy B, Hoekman L, van den Broek B, Teunissen H, Debets DO, Severson T, Jonkers J, Reddy T, de Visser KE, Faller W, Beijersbergen R, Altelaar M, de Wit E, Medema R, and Zwart W
- Subjects
- Animals, Cell Cycle genetics, Cell Line, Tumor, Cell Proliferation genetics, Cell Survival drug effects, Chromatin genetics, Chromatin Immunoprecipitation Sequencing, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Cyclin-Dependent Kinase Inhibitor p57 genetics, Enhancer Elements, Genetic, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic genetics, Humans, Imidazoles pharmacology, Immunohistochemistry, Lung Neoplasms genetics, Mice, Proteomics, Pyrazines pharmacology, RNA, Small Interfering, RNA-Seq, Receptor, IGF Type 1 metabolism, Transcription Factors genetics, Transcription Factors metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Cell Proliferation drug effects, Chromatin metabolism, Cyclin-Dependent Kinase Inhibitor p57 metabolism, Glucocorticoids pharmacology, Lung Neoplasms metabolism, Receptors, Glucocorticoid metabolism
- Abstract
The glucocorticoid receptor (GR) regulates gene expression, governing aspects of homeostasis, but is also involved in cancer. Pharmacological GR activation is frequently used to alleviate therapy-related side-effects. While prior studies have shown GR activation might also have anti-proliferative action on tumours, the underpinnings of glucocorticoid action and its direct effectors in non-lymphoid solid cancers remain elusive. Here, we study the mechanisms of glucocorticoid response, focusing on lung cancer. We show that GR activation induces reversible cancer cell dormancy characterised by anticancer drug tolerance, and activation of growth factor survival signalling accompanied by vulnerability to inhibitors. GR-induced dormancy is dependent on a single GR-target gene, CDKN1C, regulated through chromatin looping of a GR-occupied upstream distal enhancer in a SWI/SNF-dependent fashion. These insights illustrate the importance of GR signalling in non-lymphoid solid cancer biology, particularly in lung cancer, and warrant caution for use of glucocorticoids in treatment of anticancer therapy related side-effects., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
41. Duality of glucocorticoid action in cancer: tumor-suppressor or oncogene?
- Author
-
Mayayo-Peralta I, Zwart W, and Prekovic S
- Subjects
- Humans, Oncogenes, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Glucocorticoids metabolism, Glucocorticoids pharmacology, Glucocorticoids therapeutic use, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Glucocorticoid receptor (GR) is a key homeostatic regulator involved in governing immune response, neuro-integration, metabolism and lung function. In conjunction with its pivotal role in human biology, GR action is critically linked to the pathology of various disease types, including cancer. While pharmacological activation of GR has been used for the treatment of various liquid cancers, its role in solid cancers is less clearly defined and seems to be cancer-type dependent. This review focuses on the molecular aspects of GR biology, spanning the structural and functional basis of response to glucocorticoids, as well as how this transcription factor operates in cancer, including the implications in disease development, progression and drug resistance.
- Published
- 2021
- Full Text
- View/download PDF
42. Androgen and glucocorticoid receptor direct distinct transcriptional programs by receptor-specific and shared DNA binding sites.
- Author
-
Kulik M, Bothe M, Kibar G, Fuchs A, Schöne S, Prekovic S, Mayayo-Peralta I, Chung HR, Zwart W, Helsen C, Claessens F, and Meijsing SH
- Subjects
- Binding Sites, Cell Line, Tumor, Gene Expression Regulation, Humans, Protein Binding, Chromatin metabolism, DNA metabolism, Receptors, Androgen metabolism, Receptors, Glucocorticoid metabolism, Transcription Factors metabolism
- Abstract
The glucocorticoid (GR) and androgen (AR) receptors execute unique functions in vivo, yet have nearly identical DNA binding specificities. To identify mechanisms that facilitate functional diversification among these transcription factor paralogs, we studied them in an equivalent cellular context. Analysis of chromatin and sequence suggest that divergent binding, and corresponding gene regulation, are driven by different abilities of AR and GR to interact with relatively inaccessible chromatin. Divergent genomic binding patterns can also be the result of subtle differences in DNA binding preference between AR and GR. Furthermore, the sequence composition of large regions (>10 kb) surrounding selectively occupied binding sites differs significantly, indicating a role for the sequence environment in guiding AR and GR to distinct binding sites. The comparison of binding sites that are shared shows that the specificity paradox can also be resolved by differences in the events that occur downstream of receptor binding. Specifically, shared binding sites display receptor-specific enhancer activity, cofactor recruitment and changes in histone modifications. Genomic deletion of shared binding sites demonstrates their contribution to directing receptor-specific gene regulation. Together, these data suggest that differences in genomic occupancy as well as divergence in the events that occur downstream of receptor binding direct functional diversification among transcription factor paralogs., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
43. Estrogen Receptor on the move: Cistromic plasticity and its implications in breast cancer.
- Author
-
Mayayo-Peralta I, Prekovic S, and Zwart W
- Subjects
- Cell Line, Tumor, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Female, Gene Expression Regulation, Neoplastic, Humans, Receptors, Estrogen, Breast Neoplasms genetics
- Abstract
Estrogen Receptor (ERα) is a hormone-driven transcription factor, critically involved in driving tumor cell proliferation in the vast majority of breast cancers (BCas). ERα binds the genome at cis-regulatory elements, dictating the expression of a large spectrum of responsive genes in 3D genomic space. While initial reports described a rather static ERα chromatin binding repertoire, we now know that ERα DNA interactions are highly versatile, altered in breast tumor development and progression, and deviate between tumors from patients with differential outcome. Multiple cellular signaling cascades are known to impinge on ERα genomic function, changing its cistrome to retarget the receptor to other regions of the genome and reprogram its impact on breast cell biology. This review describes the current state-of-the-art on which factors manipulate the ERα cistrome and how this alters the response to both endogenous and exogenous hormonal stimuli, ultimately impacting BCa cell progression and response to commonly used therapeutic interventions. Novel insights in ERα cistrome dynamics may pave the way for better patient diagnostics and the development of novel therapeutic interventions, ultimately improving cancer care and patient outcome., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
44. Endonuclease FEN1 Coregulates ERα Activity and Provides a Novel Drug Interface in Tamoxifen-Resistant Breast Cancer.
- Author
-
Flach KD, Periyasamy M, Jadhav A, Dorjsuren D, Siefert JC, Hickey TE, Opdam M, Patel H, Canisius S, Wilson DM 3rd, Donaldson Collier M, Prekovic S, Nieuwland M, Kluin RJC, Zakharov AV, Wesseling J, Wessels LFA, Linn SC, Tilley WD, Simeonov A, Ali S, and Zwart W
- Subjects
- Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms genetics, Cell Line, Tumor, Estrogen Receptor alpha genetics, Female, Flap Endonucleases genetics, Gene Expression Regulation, Neoplastic physiology, Humans, Tamoxifen therapeutic use, Breast Neoplasms pathology, Drug Resistance, Neoplasm genetics, Estrogen Receptor alpha metabolism, Flap Endonucleases metabolism
- Abstract
Estrogen receptor α (ERα) is a key transcriptional regulator in the majority of breast cancers. ERα-positive patients are frequently treated with tamoxifen, but resistance is common. In this study, we refined a previously identified 111-gene outcome prediction-classifier, revealing FEN1 as the strongest determining factor in ERα-positive patient prognostication. FEN1 levels were predictive of outcome in tamoxifen-treated patients, and FEN1 played a causal role in ERα-driven cell growth. FEN1 impacted the transcriptional activity of ERα by facilitating coactivator recruitment to the ERα transcriptional complex. FEN1 blockade induced proteasome-mediated degradation of activated ERα, resulting in loss of ERα-driven gene expression and eradicated tumor cell proliferation. Finally, a high-throughput 465,195 compound screen identified a novel FEN1 inhibitor, which effectively blocked ERα function and inhibited proliferation of tamoxifen-resistant cell lines as well as ex vivo -cultured ERα-positive breast tumors. Collectively, these results provide therapeutic proof of principle for FEN1 blockade in tamoxifen-resistant breast cancer. SIGNIFICANCE: These findings show that pharmacologic inhibition of FEN1, which is predictive of outcome in tamoxifen-treated patients, effectively blocks ERα function and inhibits proliferation of tamoxifen-resistant tumor cells., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
45. TLE3 loss confers AR inhibitor resistance by facilitating GR-mediated human prostate cancer cell growth.
- Author
-
Palit SA, Vis D, Stelloo S, Lieftink C, Prekovic S, Bekers E, Hofland I, Šuštić T, Wolters L, Beijersbergen R, Bergman AM, Győrffy B, Wessels LF, Zwart W, and van der Heijden MS
- Subjects
- Androgen Receptor Antagonists pharmacology, Benzamides, CRISPR-Cas Systems genetics, Cell Line, Tumor, Cell Proliferation drug effects, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic drug effects, HEK293 Cells, Humans, Male, Nitriles, Phenylthiohydantoin analogs & derivatives, Phenylthiohydantoin pharmacology, Prostate metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Receptors, Glucocorticoid genetics, Transcriptional Activation drug effects, Co-Repressor Proteins genetics, Hepatocyte Nuclear Factor 3-alpha genetics, Prostatic Neoplasms genetics, Receptors, Androgen genetics
- Abstract
Androgen receptor (AR) inhibitors represent the mainstay of prostate cancer treatment. In a genome-wide CRISPR-Cas9 screen using LNCaP prostate cancer cells, loss of co-repressor TLE3 conferred resistance to AR antagonists apalutamide and enzalutamide. Genes differentially expressed upon TLE3 loss share AR as the top transcriptional regulator, and TLE3 loss rescued the expression of a subset of androgen-responsive genes upon enzalutamide treatment. GR expression was strongly upregulated upon AR inhibition in a TLE3 -negative background. This was consistent with binding of TLE3 and AR at the GR locus. Furthermore, GR binding was observed proximal to TLE3/AR-shared genes. GR inhibition resensitized TLE3
KO cells to enzalutamide. Analyses of patient samples revealed an association between TLE3 and GR levels that reflected our findings in LNCaP cells, of which the clinical relevance is yet to be determined. Together, our findings reveal a mechanistic link between TLE3 and GR-mediated resistance to AR inhibitors in human prostate cancer., Competing Interests: SP, DV, SS, CL, SP, EB, IH, TŠ, LW, RB, AB, BG, LW, Mv No competing interests declared, WZ Reviewing editor, eLife, (© 2019, Palit et al.)- Published
- 2019
- Full Text
- View/download PDF
46. A CRISPR-Cas9 screen identifies essential CTCF anchor sites for estrogen receptor-driven breast cancer cell proliferation.
- Author
-
Korkmaz G, Manber Z, Lopes R, Prekovic S, Schuurman K, Kim Y, Teunissen H, Flach K, Wit E, Galli GG, Zwart W, Elkon R, and Agami R
- Subjects
- Binding Sites genetics, Breast Neoplasms pathology, CRISPR-Cas Systems genetics, Chromatin genetics, Enhancer Elements, Genetic genetics, Female, Humans, MCF-7 Cells, Protein Binding genetics, Breast Neoplasms genetics, CCCTC-Binding Factor genetics, Cell Proliferation genetics, Estrogen Receptor alpha genetics
- Abstract
Estrogen receptor α (ERα) is an enhancer activating transcription factor, a key driver of breast cancer and a main target for cancer therapy. ERα-mediated gene regulation requires proper chromatin-conformation to facilitate interactions between ERα-bound enhancers and their target promoters. A major determinant of chromatin structure is the CCCTC-binding factor (CTCF), that dimerizes and together with cohesin stabilizes chromatin loops and forms the boundaries of topologically associated domains. However, whether CTCF-binding elements (CBEs) are essential for ERα-driven cell proliferation is unknown. To address this question in a global manner, we implemented a CRISPR-based functional genetic screen targeting CBEs located in the vicinity of ERα-bound enhancers. We identified four functional CBEs and demonstrated the role of one of them in inducing chromatin conformation changes in favor of activation of PREX1, a key ERα target gene in breast cancer. Indeed, high PREX1 expression is a bona-fide marker of ERα-dependency in cell lines, and is associated with good outcome after anti-hormonal treatment. Altogether, our data show that distinct CTCF-mediated chromatin structures are required for ERα- driven breast cancer cell proliferation., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
47. Drivers of AR indifferent anti-androgen resistance in prostate cancer cells.
- Author
-
Handle F, Prekovic S, Helsen C, Van den Broeck T, Smeets E, Moris L, Eerlings R, Kharraz SE, Urbanucci A, Mills IG, Joniau S, Attard G, and Claessens F
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Docetaxel pharmacology, Humans, Male, Mice, Mice, Nude, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Prostate drug effects, Prostate metabolism, Prostatic Neoplasms, Castration-Resistant metabolism, Signal Transduction drug effects, Androgen Antagonists pharmacology, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Prostatic Neoplasms, Castration-Resistant drug therapy, Receptors, Androgen metabolism
- Abstract
Inhibition of the androgen receptor (AR) by second-generation anti-androgens is a standard treatment for metastatic castration resistant prostate cancer (mCRPC), but it inevitably leads to the development of resistance. Since the introduction of highly efficient AR signalling inhibitors, approximately 20% of mCRPC patients develop disease with AR independent resistance mechanisms. In this study, we generated two anti-androgen and castration resistant prostate cancer cell models that do not rely on AR activity for growth despite robust AR expression (AR indifferent). They are thus resistant against all modern AR signalling inhibitors. Both cell lines display cross-resistance against the chemotherapeutic drug docetaxel due to MCL1 upregulation but remain sensitive to the PARP inhibitor olaparib and the pan-BCL inhibitor obatoclax. RNA-seq analysis of the anti-androgen resistant cell lines identified hyper-activation of the E2F cell-cycle master regulator as driver of AR indifferent growth, which was caused by deregulation of cyclin D/E, E2F1, RB1, and increased Myc activity. Importantly, mCRPC tissue samples with low AR activity displayed the same alterations and increased E2F activity. In conclusion, we describe two cellular models that faithfully mimic the acquisition of a treatment induced AR independent phenotype that is cross-resistant against chemotherapy and driven by E2F hyper-activation.
- Published
- 2019
- Full Text
- View/download PDF
48. Loss of p53 triggers WNT-dependent systemic inflammation to drive breast cancer metastasis.
- Author
-
Wellenstein MD, Coffelt SB, Duits DEM, van Miltenburg MH, Slagter M, de Rink I, Henneman L, Kas SM, Prekovic S, Hau CS, Vrijland K, Drenth AP, de Korte-Grimmerink R, Schut E, van der Heijden I, Zwart W, Wessels LFA, Schumacher TN, Jonkers J, and de Visser KE
- Subjects
- Animals, Breast Neoplasms complications, Disease Models, Animal, Female, Inflammation complications, Inflammation immunology, Interleukin-1beta immunology, Interleukin-1beta metabolism, Mice, Neutrophils immunology, Breast Neoplasms genetics, Breast Neoplasms pathology, Inflammation genetics, Inflammation pathology, Neoplasm Metastasis pathology, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Wnt Proteins metabolism
- Abstract
Cancer-associated systemic inflammation is strongly linked to poor disease outcome in patients with cancer
1,2 . For most human epithelial tumour types, high systemic neutrophil-to-lymphocyte ratios are associated with poor overall survival3 , and experimental studies have demonstrated a causal relationship between neutrophils and metastasis4,5 . However, the cancer-cell-intrinsic mechanisms that dictate the substantial heterogeneity in systemic neutrophilic inflammation between tumour-bearing hosts are largely unresolved. Here, using a panel of 16 distinct genetically engineered mouse models for breast cancer, we uncover a role for cancer-cell-intrinsic p53 as a key regulator of pro-metastatic neutrophils. Mechanistically, loss of p53 in cancer cells induced the secretion of WNT ligands that stimulate tumour-associated macrophages to produce IL-1β, thus driving systemic inflammation. Pharmacological and genetic blockade of WNT secretion in p53-null cancer cells reverses macrophage production of IL-1β and subsequent neutrophilic inflammation, resulting in reduced metastasis formation. Collectively, we demonstrate a mechanistic link between the loss of p53 in cancer cells, secretion of WNT ligands and systemic neutrophilia that potentiates metastatic progression. These insights illustrate the importance of the genetic makeup of breast tumours in dictating pro-metastatic systemic inflammation, and set the stage for personalized immune intervention strategies for patients with cancer.- Published
- 2019
- Full Text
- View/download PDF
49. Identification of mineralocorticoid receptor target genes in the mouse hippocampus.
- Author
-
van Weert LTCM, Buurstede JC, Sips HCM, Vettorazzi S, Mol IM, Hartmann J, Prekovic S, Zwart W, Schmidt MV, Roozendaal B, Tuckermann JP, Sarabdjitsingh RA, and Meijer OC
- Subjects
- Animals, Binding Sites genetics, Female, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Receptors, Mineralocorticoid genetics, Gene Expression Regulation, Hippocampus metabolism, Receptors, Mineralocorticoid physiology
- Abstract
Brain mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) respond to the same glucocorticoid hormones but can have differential effects on cellular function. Several lines of evidence suggest that MR-specific target genes must exist and might underlie the distinct effects of the receptors. The present study aimed to identify MR-specific target genes in the hippocampus, a brain region where MR and GR are co-localised and play a role in the stress response. Using genome-wide binding of both receptor types, we previously identified MR-specific, MR-GR overlapping and GR-specific putative target genes. We now report altered gene expression levels of such genes in the hippocampus of forebrain MR knockout (fbMRKO) mice, killed at the time of their endogenous corticosterone peak. Of those genes associated with MR-specific binding, the most robust effect was a 50% reduction in Jun dimerization protein 2 (Jdp2) mRNA levels in fbMRKO mice. Down-regulation was also observed for the MR-specific Nitric oxide synthase 1 adaptor protein (Nos1ap) and Suv3 like RNA helicase (Supv3 l1). Interestingly, the classical glucocorticoid target gene FK506 binding protein 5 (Fkbp5), which is associated with MR and GR chromatin binding, was expressed at substantially lower levels in fbMRKO mice. Subsequently, hippocampal Jdp2 was confirmed to be up-regulated in a restraint stress model, posing Jdp2 as a bona fide MR target that is also responsive in an acute stress condition. Thus, we show that MR-selective DNA binding can reveal functional regulation of genes and further identify distinct MR-specific effector pathways., (© 2019 The Authors. Journal of Neuroendocrinology published by John Wiley & Sons Ltd on behalf of British Society for Neuroendocrinology.)
- Published
- 2019
- Full Text
- View/download PDF
50. Enzalutamide therapy for advanced prostate cancer: efficacy, resistance and beyond
- Author
-
Linder S, van der Poel HG, Bergman AM, Zwart W, and Prekovic S
- Subjects
- Benzamides, Drug Resistance, Neoplasm drug effects, Humans, Male, Nitriles, Phenylthiohydantoin pharmacology, Phenylthiohydantoin therapeutic use, Phenylthiohydantoin analogs & derivatives, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
The androgen receptor drives the growth of metastatic castration-resistant prostate cancer. This has led to the development of multiple novel drugs targeting this hormone-regulated transcription factor, such as enzalutamide – a potent androgen receptor antagonist. Despite the plethora of possible treatment options, the absolute survival benefit of each treatment separately is limited to a few months. Therefore, current research efforts are directed to determine the optimal sequence of therapies, discover novel drugs effective in metastatic castration-resistant prostate cancer and define patient subpopulations that ultimately benefit from these treatments. Molecular studies provide evidence on which pathways mediate treatment resistance and may lead to improved treatment for metastatic castration-resistant prostate cancer. This review provides, firstly a concise overview of the clinical development, use and effectiveness of enzalutamide in the treatment of advanced prostate cancer, secondly it describes translational research addressing enzalutamide response vs resistance and lastly highlights novel potential treatment strategies in the enzalutamide-resistant setting.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.