Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Cell Sci ; 135(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35713120

ABSTRACT

Stress granules (SGs) can assemble in cancer cells upon chemotoxic stress. Glucocorticoids function during stress responses and are administered with chemotherapies. The roles of glucocorticoids in SG assembly and disassembly pathways are unknown. We examined whether combining glucocorticoids such as cortisone with chemotherapies from the vinca alkaloid family, which dismantle the microtubule network, affects SG assembly and disassembly pathways and influences cell viability in cancer cells and human-derived organoids. Cortisone augmented SG formation when combined with vinorelbine (VRB). Live-cell imaging showed that cortisone increased SG assembly rates but reduced SG clearance rates after stress, by increasing protein residence times within the SGs. Mechanistically, VRB and cortisone signaled through the integrated stress response mediated by eIF2α (also known as EIF2S1), yet induced different kinases, with cortisone activating the GCN2 kinase (also known as EIF2AK4). Cortisone increased VRB-induced cell death and reduced the population of cells trapped in mitotic catastrophe. These effects were mediated by the core SG proteins G3BP1 and G3BP2. In conclusion, glucocorticoids induce SG assembly and cell death when administered with chemotherapies, suggesting that combining glucocorticoids with chemotherapies can enhance cancer cell chemosensitivity.


Subject(s)
Cortisone , Glucocorticoids , Cell Death , Cortisone/metabolism , Cytoplasmic Granules/metabolism , DNA Helicases , Glucocorticoids/pharmacology , Humans , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Serine-Threonine Kinases , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Stress Granules
2.
PLoS Comput Biol ; 18(11): e1010584, 2022 11.
Article in English | MEDLINE | ID: mdl-36350878

ABSTRACT

Organoids have immense potential as ex vivo disease models for drug discovery and personalized drug screening. Dynamic changes in individual organoid morphology, number, and size can indicate important drug responses. However, these metrics are difficult and labor-intensive to obtain for high-throughput image datasets. Here, we present OrganoID, a robust image analysis platform that automatically recognizes, labels, and tracks single organoids, pixel-by-pixel, in brightfield and phase-contrast microscopy experiments. The platform was trained on images of pancreatic cancer organoids and validated on separate images of pancreatic, lung, colon, and adenoid cystic carcinoma organoids, which showed excellent agreement with manual measurements of organoid count (95%) and size (97%) without any parameter adjustments. Single-organoid tracking accuracy remained above 89% over a four-day time-lapse microscopy study. Automated single-organoid morphology analysis of a chemotherapy dose-response experiment identified strong dose effect sizes on organoid circularity, solidity, and eccentricity. OrganoID enables straightforward, detailed, and accurate image analysis to accelerate the use of organoids in high-throughput, data-intensive biomedical applications.


Subject(s)
Deep Learning , Organoids , Colon , Drug Discovery , High-Throughput Nucleotide Sequencing
3.
PLoS Genet ; 15(11): e1008459, 2019 11.
Article in English | MEDLINE | ID: mdl-31765392

ABSTRACT

Gene expression dynamics can be measured in single living cells. Using a detectable transcriptionally active gene in living cells, we previously found that an mRNA undergoing several splicing events was retained at this gene after transcription until completion of mRNA processing. To determine the reason for this delay in release and whether mRNA retention on the gene might depend on splicing factor availability, we modulated the levels of splicing factors in the nucleus. Increasing the abundance of the diffusing fraction of splicing factors by their overexpression or by Clk1 kinase overexpression to disassemble nuclear speckles, led to a reduction in splicing factor residence times on the active gene, and the retained mRNA was rapidly released from the gene. Other treatments such as overexpression of a mutant inactive Clk1, the downregulation of MALAT1 lncRNA or of the Son protein, or the overexpression of the splicing factor import factor TNPO3, did not affect the dynamics of mRNA release from the gene. We found that the faster release of the mRNA from the gene mediated by increased availability of splicing factors, was dependent on the RS domain of the splicing factors and its phosphorylation state. We propose that the relative abundancies of splicing factors in the nucleoplasm can affect their availability for the splicing events taking place, and regulate the kinetics of mRNA release from the gene after processing.


Subject(s)
RNA Splicing Factors/genetics , RNA Splicing/genetics , Transcription, Genetic , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , HeLa Cells , Humans , Introns/genetics , Minor Histocompatibility Antigens/genetics , Phosphorylation , Protein Binding/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , RNA Precursors/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , beta Karyopherins/genetics
4.
Trends Genet ; 32(7): 419-431, 2016 07.
Article in English | MEDLINE | ID: mdl-27185238

ABSTRACT

Transcribed mRNA molecules must reach the cytoplasm to undergo translation. Technological developments in imaging have placed mRNAs under the spotlight, allowing the quantitative study of the spatial and temporal dynamics of the nucleocytoplasmic mRNA export process. Here, we discuss studies that have used such experimental approaches to demonstrate that gene tethering at the nuclear pore complex (NPC) regulates mRNA expression, and to characterize mRNA dynamics during transport in real time. The paths taken by mRNAs as they move from their sites of transcription and travel through the nucleoplasm, in between chromatin domains, and finally through the NPC, can now be observed in detail.


Subject(s)
Active Transport, Cell Nucleus/genetics , Nuclear Pore/genetics , RNA, Messenger/genetics , Transcription, Genetic , Cell Nucleus/genetics , Chromatin/genetics , Cytoplasm/genetics , Gene Expression Regulation , RNA Transport/genetics , Ribonucleoproteins/genetics
5.
Genes Dev ; 25(10): 1052-64, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21576265

ABSTRACT

Nuclear export of messenger RNA (mRNA) occurs by translocation of mRNA/protein complexes (mRNPs) through nuclear pore complexes (NPCs). The DEAD-box protein Dbp5 mediates export by triggering removal of mRNP proteins in a spatially controlled manner. This requires Dbp5 interaction with Nup159 in NPC cytoplasmic filaments and activation of Dbp5's ATPase activity by Gle1 bound to inositol hexakisphosphate (IP(6)). However, the precise sequence of events within this mechanism has not been fully defined. Here we analyze dbp5 mutants that alter ATP binding, ATP hydrolysis, or RNA binding. We found that ATP binding and hydrolysis are required for efficient Dbp5 association with NPCs. Interestingly, mutants defective for RNA binding are dominant-negative (DN) for mRNA export in yeast and human cells. We show that the DN phenotype stems from competition with wild-type Dbp5 for Gle1 at NPCs. The Dbp5-Gle1 interaction is limiting for export and, importantly, can be independent of Nup159. Fluorescence recovery after photobleaching experiments in yeast show a very dynamic association between Dbp5 and NPCs, averaging <1 sec, similar to reported NPC translocation rates for mRNPs. This work reveals critical steps in the Gle1-IP(6)/Dbp5/Nup159 cycle, and suggests that the number of remodeling events mediated by a single Dbp5 is limited.


Subject(s)
Cell Nucleus/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus , Adenosine Triphosphate/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Hydrolysis , Mutation , Nuclear Pore Complex Proteins/metabolism , Phenotype , Protein Binding/genetics , Saccharomyces cerevisiae/growth & development
6.
J Cell Sci ; 127(Pt 20): 4443-56, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128566

ABSTRACT

The 5'-to-3' mRNA degradation machinery localizes to cytoplasmic processing bodies (P-bodies), which are non-membranous structures found in all eukaryotes. Although P-body function has been intensively studied in yeast, less is known about their role in mammalian cells, such as whether P-body enzymes are actively engaged in mRNA degradation or whether P-bodies serve as mRNA storage depots, particularly during cellular stress. We examined the fate of mammalian mRNAs in P-bodies during translational stress, and show that mRNAs accumulate within P-bodies during amino acid starvation. The 5' and 3' ends of the transcripts residing in P-bodies could be identified, but poly(A) tails were not detected. Using the MS2 mRNA-tagging system for mRNA visualization in living cells, we found that a stationary mRNA population formed in P-bodies during translational stress, which cleared gradually after the stress was relieved. Dcp2-knockdown experiments showed that there is constant degradation of part of the P-body-associated mRNA population. This analysis demonstrates the dual role of P-bodies as decay sites and storage areas under regular and stress conditions.


Subject(s)
Cellular Structures/metabolism , Cytoplasm/metabolism , Endoribonucleases/metabolism , Protein Biosynthesis , RNA Stability , RNA, Messenger/genetics , Amino Acids/deficiency , Cell Line , Endoribonucleases/genetics , Gene Knockdown Techniques , Humans , Stress, Physiological/genetics , Time-Lapse Imaging
7.
Cells ; 13(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39273076

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays a major role in breast cancer progression and the development of drug resistance. We have previously demonstrated a trans-differentiation therapeutic approach targeting invasive dedifferentiated cancer cells. Using a combination of PPARγ agonists and MEK inhibitors, we forced the differentiation of disseminating breast cancer cells into post-mitotic adipocytes. Utilizing murine breast cancer cells, we demonstrated a broad class effect of PPARγ agonists and MEK inhibitors in inducing cancer cell trans-differentiation into adipocytes. Both Rosiglitazone and Pioglitazone effectively induced adipogenesis in cancer cells, marked by PPARγ and C/EBPα upregulation, cytoskeleton rearrangement, and lipid droplet accumulation. All tested MEK inhibitors promoted adipogenesis in the presence of TGFß, with Cobimetinib showing the most prominent effects. A metastasis ex vivo culture from a patient diagnosed with triple-negative breast cancer demonstrated a synergistic upregulation of PPARγ with the combination of Pioglitazone and Cobimetinib. Our results highlight the potential for new therapeutic strategies targeting cancer cell plasticity and the dedifferentiation phenotype in aggressive breast cancer subtypes. Combining differentiation treatments with standard therapeutic approaches may offer a strategy to overcome drug resistance.


Subject(s)
Cell Differentiation , PPAR gamma , Pioglitazone , PPAR gamma/metabolism , PPAR gamma/agonists , Humans , Animals , Mice , Cell Differentiation/drug effects , Cell Line, Tumor , Female , Pioglitazone/pharmacology , Protein Kinase Inhibitors/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Epithelial-Mesenchymal Transition/drug effects , Rosiglitazone/pharmacology , Azetidines/pharmacology , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Piperidines/pharmacology
8.
Sci Rep ; 13(1): 20513, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37993670

ABSTRACT

Disruption of intestinal epithelial functions is linked to Crohn disease (CD) pathogenesis. We identified a widespread reduction in the expression of long non-coding RNAs (lncRNAs) including LHFPL3-AS2 in the treatment-naïve CD ileum of the RISK pediatric cohort. We validated the reduction of LHFPL3-AS2 in adult CD and noted a further reduction in patients with more severe CD from the RISK cohort. LHFPL3-AS2 knockdown in Caco-2 cells robustly affected epithelial monolayer morphogenesis with markedly reduced confluency and spreading, showing atypical rounding, and clumping. mRNA-seq analysis of LHFPL3-AS2 knockdown cells highlighted the reduction of genes and pathways linked with apical polarity, actin bundles, morphogenesis, and the b-catenin-TCF4 complex. LHFPL3-AS2 knockdown significantly reduced the ability of cells to form an internal lumen within the 3-dimensional (3D) cyst model, with mislocalization of actin and adherent and tight junction proteins, affecting epithelial polarity. LHFPL3-AS2 knockdown also resulted in defective mitotic spindle formation and consequent reduction in epithelial proliferation. Altogether, we show that LHFPL3-AS2 reduction affects epithelial morphogenesis, polarity, mitotic spindle formation, and proliferation, which are key processes in maintaining epithelial homeostasis in CD. Reduced expression of LHFPL3-AS2 in CD patients and its further reduction with ileal ulceration outcome, emphasizes its significance in this context.


Subject(s)
Crohn Disease , RNA, Long Noncoding , Adult , Humans , Child , Caco-2 Cells , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Crohn Disease/genetics , Actins/genetics , Cell Proliferation/genetics , Ileum/metabolism , Cell Line, Tumor
9.
J Crohns Colitis ; 17(6): 960-971, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-36655602

ABSTRACT

BACKGROUND AND AIMS: Widespread dysregulation of long non-coding RNAs [lncRNAs] including a reduction in GATA6-AS1 was noted in inflammatory bowel disease [IBD]. We previously reported a prominent inhibition of epithelial mitochondrial functions in ulcerative colitis [UC]. However, the connection between reduction of GATA6-AS1 expression and attenuated epithelial mitochondrial functions was not defined. METHODS: Mucosal transcriptomics was used to conform GATA6-AS1 reduction in several treatment-naïve independent human cohorts [n=673]. RNA pull-down followed by mass spectrometry was used to determine the GATA6-AS1 interactome. Metabolomics and mitochondrial respiration following GATA6-AS1 silencing in Caco-2 cells were used to elaborate on GATA6-AS1 functions. RESULTS: GATA6-AS1 showed predominant expression in gut epithelia using single cell datasets. GATA6-AS1 levels were reduced in Crohn's disease [CD] ileum and UC rectum in independent cohorts. Reduced GATA6-AS1 lncRNA was further linked to a more severe UC form, and to a less favourable UC course. The GATA6-AS1 interactome showed robust enrichment for mitochondrial proteins, and included TGM2, an autoantigen in coeliac disease that is induced in UC, CD and coeliac disease, in contrast to GATA6-AS1 reduction in these cohorts. GATA6-AS1 silencing resulted in induction of TGM2, and this was coupled with a reduction in mitochondrial membrane potential and mitochondrial respiration, as well as in a reduction of metabolites linked to aerobic respiration relevant to mucosal inflammation. TGM2 knockdown in GATA6-AS1-deficient cells rescued mitochondrial respiration. CONCLUSIONS: GATA6-AS1 levels are reduced in UC, CD and coeliac disease, and in more severe UC forms. We highlight GATA6-AS1 as a target regulating epithelial mitochondrial functions, potentially through controlling TGM2 levels.


Subject(s)
Celiac Disease , Colitis, Ulcerative , Crohn Disease , Humans , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Caco-2 Cells , Intestinal Mucosa/metabolism , Crohn Disease/metabolism , Rectum , Inflammation/metabolism , Mitochondria/metabolism , GATA6 Transcription Factor/metabolism
10.
Curr Opin Cell Biol ; 58: 69-75, 2019 06.
Article in English | MEDLINE | ID: mdl-30889416

ABSTRACT

The mRNA molecule roams through the nucleus on its way out to the cytoplasm. mRNA encounters and is bound by many protein factors, from the moment it begins to emerge from RNA polymerase II and during its travel in the nucleoplasm, where it will come upon chromatin and nuclear bodies. Some of the protein factors that engage with the mRNA can process it, until finally reaching a mature state fit for export through the nuclear pore complex (NPC). Examining the lifecycle of mRNAs in living cells using mRNA tagging techniques opens a window into our understanding of the rules that drive the dynamics of gene expression from transcription to mRNA export.


Subject(s)
Cell Nucleus/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Chromatin/metabolism , Cytoplasm/metabolism , Humans , Nuclear Pore/metabolism , RNA Polymerase II/metabolism , RNA Stability , RNA Transport , Transcription, Genetic
11.
Methods Mol Biol ; 2038: 151-163, 2019.
Article in English | MEDLINE | ID: mdl-31407283

ABSTRACT

Export of mRNA transcripts from the cell nucleus is a complex and multistep process, regulated by various proteins and control mechanisms. Recent studies have demonstrated the rapid passage of mRNA-protein complexes (mRNPs) through the nuclear pore complex (NPC) as well as the ability to detect mRNPs stalled at the NPC during inhibition of the mRNA export process. In this chapter, we describe ways to block mRNA export and present an image analysis method to identify mRNPs stuck at the NPC during such blocks. Using the MS2 mRNA-tagging system to track single mRNPs in living cells we are able to examine their intracellular distribution and dynamics both in the nucleoplasm and at the nuclear periphery. We use this method to identify and count the number of static mRNPs anchored to the nuclear envelope under different conditions of mRNA export inhibition.


Subject(s)
Microscopy, Fluorescence , Molecular Imaging/methods , Nuclear Envelope/metabolism , RNA, Messenger/metabolism , Single Molecule Imaging/methods , Active Transport, Cell Nucleus , Animals , Cells, Cultured , Gene Expression Regulation , Humans , Nuclear Envelope/genetics , RNA, Messenger/genetics , Time Factors
12.
J Cell Biol ; 218(9): 2962-2981, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31375530

ABSTRACT

Translocation of mRNA through the nuclear pore complex (NPC) requires interactions with different NPC regions. To determine the interactions that are crucial for effective mRNA export in living cells, we examined mRNA export within individual pores by applying various types of mRNA export blocks that stalled mRNPs at different stages of transition. Focusing on the major mRNA export factor NXF1, we found that initial mRNP binding to the NPC did not require NXF1 in the NPC, whereas release into the cytoplasm did. NXF1 localization in the NPC did not require RNA or RNA binding. Superresolution microscopy showed that NXF1 consistently occupied positions on the cytoplasmic side of the NPC. Interactions with specific nucleoporins were pinpointed using FLIM-FRET for measuring protein-protein interactions inside single NPCs, showing that Dbp5 helicase activity of mRNA release is conserved in yeast and humans. Altogether, we find that specific interactions on the cytoplasmic side of the NPC are fundamental for the directional flow of mRNA export.


Subject(s)
Cytoplasm/metabolism , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Cell Line, Tumor , Cytoplasm/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Humans , Nuclear Pore/genetics , Nucleocytoplasmic Transport Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
13.
Nucleus ; 1(6): 492-8, 2010.
Article in English | MEDLINE | ID: mdl-21327092

ABSTRACT

The transcription machinery in the eukaryotic nucleus generates messenger RNA molecules that translocate through the nucleoplasm, anchor to a nuclear pore, and find their way out into the cytoplasm. The dynamic aspects of these steps in the expression pathway were examined in order to understand the kinetic time-frames of gene activation and message dissemination. Utilizing live-cell imaging and tracking of single mRNPs containing different sized mRNAs and varying numbers of introns and exons, it was possible to quantify the temporal and spatial characteristics of the nucleoplasmic travels of mRNPs as well as the kinetics of translocation through the nuclear pore.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Animals , Cells, Cultured , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Transcription, Genetic , Wheat Germ Agglutinins/metabolism
14.
Nat Cell Biol ; 12(6): 543-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20453848

ABSTRACT

The flow of genetic information in eukaryotic cells occurs through the nucleocytoplasmic translocation of mRNAs. Knowledge of in vivo messenger RNA export kinetics remains poor in comparison with that of protein transport. We have established a mammalian system that allowed the real-time visualization and quantification of large single mRNA-protein complexes (mRNPs) during export. The in vivo dynamics of bulk mRNP transport and export, from transcription to the nuclear pore complex (NPC), occurred within a 5-40 minute time frame, with no NPC pile-up. mRNP export was rapid (about 0.5 s) and kinetically faster than nucleoplasmic diffusion. Export inhibition demonstrated that mRNA-NPC interactions were independent of ongoing export. Nucleoplasmic transport dynamics of intron-containing and intronless mRNAs were similar, yet an intron did increase export efficiency. Here we provide visualization and analysis at the single mRNP level of the various steps in nuclear gene expression and the inter-chromatin tracks through which mRNPs diffuse, and demonstrate the kinetics of mRNP-NPC interactions and translocation.


Subject(s)
Cell Nucleus/metabolism , Cells/metabolism , Nuclear Pore/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Biological Transport/genetics , Cell Nucleus/genetics , Chromosomes/metabolism , Introns , Mammals/genetics , Mammals/metabolism , Nuclear Pore/genetics , Protein Transport/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL