Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
Front Mol Biosci ; 10: 1187187, 2023.
Article En | MEDLINE | ID: mdl-37228587

Cells respond to inflammatory stimuli such as cytokines by activation of the nuclear factor-κB (NF-κB) signalling pathway, resulting in oscillatory translocation of the transcription factor p65 between nucleus and cytoplasm in some cell types. We investigate the relationship between p65 and inhibitor-κB⍺ (IκBα) protein levels and dynamic properties of the system, and how this interaction impacts on the expression of key inflammatory genes. Using bacterial artificial chromosomes, we developed new cell models of IκB⍺-eGFP protein overexpression in a pseudo-native genomic context. We find that cells with high levels of the negative regulator IκBα remain responsive to inflammatory stimuli and maintain dynamics for both p65 and IκBα. In contrast, canonical target gene expression is dramatically reduced by overexpression of IκBα, but can be partially rescued by overexpression of p65. Treatment with leptomycin B to promote nuclear accumulation of IκB⍺ also suppresses canonical target gene expression, suggesting a mechanism in which nuclear IκB⍺ accumulation prevents productive p65 interaction with promoter binding sites. This causes reduced target promoter binding and gene transcription, which we validate by chromatin immunoprecipitation and in primary cells. Overall, we show how inflammatory gene transcription is modulated by the expression levels of both IκB⍺ and p65. This results in an anti-inflammatory effect on transcription, demonstrating a broad mechanism to modulate the strength of inflammatory response.

2.
Cell Syst ; 11(3): 300-314.e8, 2020 09 23.
Article En | MEDLINE | ID: mdl-32918862

Single-cell gene expression is inherently variable, but how this variability is controlled in response to stimulation remains unclear. Here, we use single-cell RNA-seq and single-molecule mRNA counting (smFISH) to study inducible gene expression in the immune toll-like receptor system. We show that mRNA counts of tumor necrosis factor α conform to a standard stochastic switch model, while transcription of interleukin-1ß involves an additional regulatory step resulting in increased heterogeneity. Despite different modes of regulation, systematic analysis of single-cell data for a range of genes demonstrates that the variability in transcript count is linearly constrained by the mean response over a range of conditions. Mathematical modeling of smFISH counts and experimental perturbation of chromatin state demonstrates that linear constraints emerge through modulation of transcriptional bursting along with gene-specific relationships. Overall, our analyses demonstrate that the variability of the inducible single-cell mRNA response is constrained by transcriptional bursting.


RNA, Messenger/genetics , Toll-Like Receptors/metabolism , Humans , Models, Theoretical , Signal Transduction
3.
Front Immunol ; 10: 2168, 2019.
Article En | MEDLINE | ID: mdl-31572379

The heterogeneous nature of inflammatory bowel disease (IBD) presents challenges, particularly when choosing therapy. Activation of the NF-κB transcription factor is a highly regulated, dynamic event in IBD pathogenesis. Using a lentivirus approach, NF-κB-regulated luciferase was expressed in patient macrophages, isolated from frozen peripheral blood mononuclear cell samples. Following activation, samples could be segregated into three clusters based on the NF-κB-regulated luciferase response. The ulcerative colitis (UC) samples appeared only in the hypo-responsive Cluster 1, and in Cluster 2. Conversely, Crohn's disease (CD) patients appeared in all Clusters with their percentage being higher in the hyper-responsive Cluster 3. A positive correlation was seen between NF-κB-induced luciferase activity and the concentrations of cytokines released into medium from stimulated macrophages, but not with serum or biopsy cytokine levels. Confocal imaging of lentivirally-expressed p65 activation revealed that a higher proportion of macrophages from CD patients responded to endotoxin lipid A compared to controls. In contrast, cells from UC patients exhibited a shorter duration of NF-κB p65 subunit nuclear localization compared to healthy controls, and CD donors. Analysis of macrophage cytokine responses and patient metadata revealed a strong correlation between CD patients who smoked and hyper-activation of p65. These in vitro dynamic assays of NF-κB activation in blood-derived macrophages have the potential to segregate IBD patients into groups with different phenotypes and may therefore help determine response to therapy.


Cell Nucleus/immunology , Colitis, Ulcerative/immunology , Crohn Disease/immunology , Macrophages/immunology , Signal Transduction/immunology , Transcription Factor RelA/immunology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Adult , Animals , Cell Nucleus/genetics , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Female , Humans , Macrophages/pathology , Male , Mice , Mice, Knockout , Middle Aged , Signal Transduction/genetics , Transcription Factor RelA/genetics
4.
Cancer Cell ; 35(3): 519-533.e8, 2019 03 18.
Article En | MEDLINE | ID: mdl-30889383

Inhibitors of poly(ADP-ribose) polymerase (PARP) have demonstrated efficacy in women with BRCA-mutant ovarian cancer. However, only 15%-20% of ovarian cancers harbor BRCA mutations, therefore additional therapies are required. Here, we show that a subset of ovarian cancer cell lines and ex vivo models derived from patient biopsies are sensitive to a poly(ADP-ribose) glycohydrolase (PARG) inhibitor. Sensitivity is due to underlying DNA replication vulnerabilities that cause persistent fork stalling and replication catastrophe. PARG inhibition is synthetic lethal with inhibition of DNA replication factors, allowing additional models to be sensitized by CHK1 inhibitors. Because PARG and PARP inhibitor sensitivity are mutually exclusive, our observations demonstrate that PARG inhibitors have therapeutic potential to complement PARP inhibitor strategies in the treatment of ovarian cancer.


DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Ovarian Neoplasms/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , Female , Glycoside Hydrolases/antagonists & inhibitors , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinazolinones/pharmacology
5.
Sci Signal ; 11(540)2018 07 24.
Article En | MEDLINE | ID: mdl-30042130

Toll-like receptor (TLR) signaling regulates macrophage activation and effector cytokine propagation in the constrained environment of a tissue. In macrophage populations, TLR4 stimulates the dose-dependent transcription of nuclear factor κB (NF-κB) target genes. However, using single-RNA counting, we found that individual cells exhibited a wide range (three orders of magnitude) of expression of the gene encoding the proinflammatory cytokine tumor necrosis factor-α (TNF-α). The TLR4-induced TNFA transcriptional response correlated with the extent of NF-κB signaling in the cells and their size. We compared the rates of TNF-α production and uptake in macrophages and mouse embryonic fibroblasts and generated a mathematical model to explore the heterogeneity in the response of macrophages to TLR4 stimulation and the propagation of the TNF-α signal in the tissue. The model predicts that the local propagation of the TLR4-dependent TNF-α response and cellular NF-κB signaling are limited to small distances of a few cell diameters between neighboring tissue-resident macrophages. In our predictive model, TNF-α propagation was constrained by competitive uptake of TNF-α from the environment, rather than by heterogeneous production of the cytokine. We propose that the highly constrained architecture of tissues enables effective localized propagation of inflammatory cues while avoiding out-of-context responses at longer distances.


Inflammation/immunology , Macrophage Activation , Macrophages/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Inflammation/metabolism , Macrophages/immunology , Mice , Mice, Transgenic , NF-kappa B/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , RAW 264.7 Cells , Single-Cell Analysis , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/immunology
6.
J Immunol ; 199(8): 2652-2667, 2017 10 15.
Article En | MEDLINE | ID: mdl-28904128

TCR signaling pathways cooperate to activate the inducible transcription factors NF-κB, NFAT, and AP-1. In this study, using the calcium ionophore ionomycin and/or PMA on Jurkat T cells, we show that the gene expression program associated with activation of TCR signaling is closely related to specific chromatin landscapes. We find that calcium and kinase signaling cooperate to induce chromatin remodeling at ∼2100 chromatin regions, which demonstrate enriched binding motifs for inducible factors and correlate with target gene expression. We found that these regions typically function as inducible enhancers. Many of these elements contain composite NFAT/AP-1 sites, which typically support cooperative binding, thus further reinforcing the need for cooperation between calcium and kinase signaling in the activation of genes in T cells. In contrast, treatment with PMA or ionomycin alone induces chromatin remodeling at far fewer regions (∼600 and ∼350, respectively), which mostly represent a subset of those induced by costimulation. This suggests that the integration of TCR signaling largely occurs at the level of chromatin, which we propose plays a crucial role in regulating T cell activation.


Calcium/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Calcium Ionophores/immunology , Humans , Jurkat Cells , Lymphocyte Activation , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Phosphotransferases/metabolism , Receptor Cross-Talk , Signal Transduction , Transcription Factor AP-1/metabolism
7.
Curr Protoc Cell Biol ; 75: 22.21.1-22.21.16, 2017 Jun 19.
Article En | MEDLINE | ID: mdl-28627755

While a detailed understanding of chromatin dynamics is needed to explain how higher-order chromatin organization influences nuclear function, the molecular principles that regulate chromatin mobility in mammalian nuclei remain largely unknown. Here we describe experimental tools to follow chromatin dynamics by labeling DNA during S phase. Using these methods, we have found that foci labeled during early and mid/late S phase have significantly different dynamic behavior. Spatially constrained heterochromatic foci restrict long-range transformations of the chromosome territory (CT) structure while providing a structural framework on which highly mobile euchromatic foci undergo positional oscillations that drive local changes in the chromosome shape. Despite often dramatic mobility, we have demonstrated a preservation of structural integrity which ensures that DNA from neighboring CTs is not able to mix freely within the same nuclear space. Finally, other potential applications of the presented protocols are discussed. © 2017 by John Wiley & Sons, Inc.


Chromosomes/ultrastructure , DNA Replication , DNA/analysis , Microscopy, Confocal/methods , Optical Imaging/methods , S Phase , Animals , Bromodeoxyuridine/analysis , Cell Culture Techniques/methods , Cell Line , Cell Tracking/methods , Chickens , Chromosomes/chemistry , Humans , Staining and Labeling/methods
8.
Nat Commun ; 7: 12057, 2016 07 06.
Article En | MEDLINE | ID: mdl-27381163

Cells respond dynamically to pulsatile cytokine stimulation. Here we report that single, or well-spaced pulses of TNFα (>100 min apart) give a high probability of NF-κB activation. However, fewer cells respond to shorter pulse intervals (<100 min) suggesting a heterogeneous refractory state. This refractory state is established in the signal transduction network downstream of TNFR and upstream of IKK, and depends on the level of the NF-κB system negative feedback protein A20. If a second pulse within the refractory phase is IL-1ß instead of TNFα, all of the cells respond. This suggests a mechanism by which two cytokines can synergistically activate an inflammatory response. Gene expression analyses show strong correlation between the cellular dynamic response and NF-κB-dependent target gene activation. These data suggest that refractory states in the NF-κB system constitute an inherent design motif of the inflammatory response and we suggest that this may avoid harmful homogenous cellular activation.


Interleukin-1beta/pharmacology , NF-KappaB Inhibitor alpha/genetics , NF-kappa B/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Luminescent Proteins/genetics , Luminescent Proteins/immunology , NF-KappaB Inhibitor alpha/immunology , NF-kappa B/immunology , Neurons , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/antagonists & inhibitors , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Red Fluorescent Protein
9.
Elife ; 52016 05 17.
Article En | MEDLINE | ID: mdl-27185527

Dynamic cellular systems reprogram gene expression to ensure appropriate cellular fate responses to specific extracellular cues. Here we demonstrate that the dynamics of Nuclear Factor kappa B (NF-κB) signalling and the cell cycle are prioritised differently depending on the timing of an inflammatory signal. Using iterative experimental and computational analyses, we show physical and functional interactions between NF-κB and the E2 Factor 1 (E2F-1) and E2 Factor 4 (E2F-4) cell cycle regulators. These interactions modulate the NF-κB response. In S-phase, the NF-κB response was delayed or repressed, while cell cycle progression was unimpeded. By contrast, activation of NF-κB at the G1/S boundary resulted in a longer cell cycle and more synchronous initial NF-κB responses between cells. These data identify new mechanisms by which the cellular response to stress is differentially controlled at different stages of the cell cycle.


Cell Cycle , Cell Proliferation , E2F1 Transcription Factor/metabolism , E2F4 Transcription Factor/metabolism , Immunity, Innate , NF-kappa B/metabolism , Signal Transduction , Cell Line , Humans
10.
Cell Cycle ; 15(3): 345-56, 2016.
Article En | MEDLINE | ID: mdl-26818565

Defects in nuclear architecture occur in a variety of diseases, however the fundamental mechanisms that control the internal structure of nuclei are poorly defined. Here we reveal that the cellular microenvironment has a profound influence on the global internal organization of nuclei in breast epithelia. A 3D microenvironment induces a prolonged but reversible form of cell cycle arrest that features many of the classical markers of cell senescence. This unique form of arrest is dependent on signaling from the external microenvironment through ß1-integrins. It is concomitant with alterations in nuclear architecture that characterize the withdrawal from cell proliferation. Unexpectedly, following prolonged cell cycle arrest in 3D, the senescence-like state and associated reprogramming of nuclear architecture are freely reversible on altering the dimensionality of the cellular microenvironment. Breast epithelia can therefore maintain a proliferative plasticity that correlates with nuclear remodelling. However, the changes in nuclear architecture are cell lineage-specific and do not occur in fibroblasts, and moreover they are overcome in breast cancer cells.


Cellular Microenvironment , Integrin beta1/metabolism , Animals , Aphidicolin/pharmacology , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Immunoblotting , MCF-7 Cells , Mammary Glands, Animal/cytology , Mice , Microscopy, Confocal
12.
PLoS One ; 8(10): e77015, 2013.
Article En | MEDLINE | ID: mdl-24130828

Induction of genes is rarely an isolated event; more typically occurring as part of a web of parallel interactions, or motifs, which act to refine and control gene expression. Here, we define an Incoherent Feed-forward Loop motif in which TNFα-induced NF-κB signalling activates expression of the TNFA gene itself and also controls synthesis of the negative regulator BCL-3. While sharing a common inductive signal, the two genes have distinct temporal expression profiles. Notably, while the TNFA gene promoter is primed to respond immediately to activated NF-κB in the nucleus, induction of BCL3 expression only occurs after a time delay of about 1h. We show that this time delay is defined by remodelling of the BCL3 gene promoter, which is required to activate gene expression, and characterise the chromatin delayed induction of BCL3 expression using mathematical models. The models show how a delay in inhibitor production effectively uncouples the rate of response to inflammatory cues from the final magnitude of inhibition. Hence, within this regulatory motif, a delayed (incoherent) feed-forward loop together with differential rates of TNFA (fast) and BCL3 (slow) mRNA turnover provide robust, pulsatile expression of TNFα . We propose that the structure of the BCL-3-dependent regulatory motif has a beneficial role in modulating expression dynamics and the inflammatory response while minimising the risk of pathological hyper-inflammation.


Chromatin/genetics , Gene Expression Regulation , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics , B-Cell Lymphoma 3 Protein , Cell Line, Tumor , Humans , Kinetics , Models, Genetic , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Transcription, Genetic , Transcriptional Activation
13.
Chromosome Res ; 21(1): 87-100, 2013 Mar.
Article En | MEDLINE | ID: mdl-23463495

Recently, a novel DNA replication precursor analogue called 5-ethynyl-2'-deoxyuridine (EdU) has been widely used to monitor DNA synthesis as an alternative to bromodeoxyuridine. Use of EdU benefits from simplicity and reproducibility and the simple chemical detection systems allows excellent preservation of nuclear structure. However, the alkyne moiety is highly reactive, raising the possibility that incorporation might compromise genome stability. To assess the extent of possible DNA damage, we have analysed the effect of EdU incorporation into DNA during short- and long-term cell culture using a variety of cell lines. We show that EdU incorporation has no measurable impact on the rate of elongation of replication forks during synthesis. However, using different cell lines we find that during long-term cell culture variable responses to EdU incorporation are seen, which range from delayed cell cycle progression to complete cell cycle arrest. The most profound phenotypes were seen in mouse embryonic stem cells, which following incorporation of EdU accumulated in the G2/M-phase of the cell cycle before undergoing apoptosis. In long-term cell culture, EdU incorporation also triggered a DNA damage response in all cell types analysed. Our study shows that while EdU is extremely useful to tag sites of on-going replication, for long-term studies (i.e. beyond the cell cycle in which labelling is performed), a careful analysis of cell cycle perturbations must be performed in order to ensure that any conclusions made after EdU treatment are not a direct consequence of EdU-dependent activation of cell stress responses.


DNA Damage/drug effects , DNA Replication/drug effects , DNA/genetics , Deoxyuridine/analogs & derivatives , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Click Chemistry , DNA/drug effects , DNA Damage/genetics , DNA Replication/genetics , Deoxyuridine/pharmacology , Flow Cytometry , Mice
14.
Methods ; 57(2): 140-8, 2012 Jun.
Article En | MEDLINE | ID: mdl-22683305

The precise regulation of DNA replication is fundamental to the preservation of intact genomes during cell proliferation. Our understanding of this process has been based traditionally on a combination of techniques including biochemistry, molecular biology and cell biology. In this report we describe how the analysis of the S phase in mammalian cells using classical cell biology techniques has contributed to our understanding of the replication process. We describe traditional and state-of-the-art protocols for imaging sites of DNA synthesis in nuclei and the organisation of active replicons along DNA, as visualised on individual DNA fibres. We evaluate how the different approaches inform our understanding of the replication process, placing particular emphasis on ways in which the higher order chromatin structures and the spatial architecture of replication sites contribute to the orderly activation of defined regions of the genome at precise times of S phase.


Chromosomes , DNA Replication , DNA/genetics , Replicon , Animals , DNA/biosynthesis , DNA/chemistry , Fixatives/chemistry , Fluorescent Antibody Technique, Indirect , Formaldehyde/chemistry , HeLa Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Immunoelectron , Polymers/chemistry , S Phase Cell Cycle Checkpoints , Single-Cell Analysis , Staining and Labeling , Tissue Fixation
15.
Proc Natl Acad Sci U S A ; 109(5): 1524-9, 2012 Jan 31.
Article En | MEDLINE | ID: mdl-22307607

Filamin A (FLNA) is an actin-binding protein with a well-established role in the cytoskeleton, where it determines cell shape and locomotion by cross-linking actin filaments. Mutations in FLNA are associated with a wide range of genetic disorders. Here we demonstrate a unique role for FLNA as a nucleolar protein that associates with the RNA polymerase I (Pol I) transcription machinery to suppress rRNA gene transcription. We show that depletion of FLNA by siRNAs increased rRNA expression, rDNA promoter activity and cell proliferation. Immunodepletion of FLNA from nuclear extracts resulted in a decrease in rDNA promoter-driven transcription in vitro. FLNA coimmunoprecipitated with the Pol I components actin, TIF-IA, and RPA40, and their occupancy of the rDNA promoter was increased in the absence of FLNA in vivo. The FLNA actin-binding domain is essential for the suppression of rRNA expression and for inhibiting recruitment of the Pol I machinery to the rDNA promoter. These findings reveal an additional role for FLNA as a regulator of rRNA gene expression and have important implications for our understanding of the role of FLNA in human disease.


Contractile Proteins/physiology , Cytoskeletal Proteins/physiology , Microfilament Proteins/physiology , RNA, Ribosomal/genetics , Transcription, Genetic/physiology , Actins , Binding Sites , Cell Line , Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , DNA, Ribosomal/genetics , Filamins , Humans , Microfilament Proteins/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering
16.
PLoS One ; 6(12): e27527, 2011.
Article En | MEDLINE | ID: mdl-22205925

The distribution of chromatin within the mammalian nucleus is constrained by its organization into chromosome territories (CTs). However, recent studies have suggested that promiscuous intra- and inter-chromosomal interactions play fundamental roles in regulating chromatin function and so might define the spatial integrity of CTs. In order to test the extent of DNA mixing between CTs, DNA foci of individual CTs were labeled in living cells following incorporation of Alexa-488 and Cy-3 conjugated replication precursor analogues during consecutive cell cycles. Uniquely labeled chromatin domains, resolved following random mitotic segregation, were visualized as discrete structures with defined borders. At the level of resolution analysed, evidence for mixing of chromatin from adjacent domains was only apparent within the surface volumes where neighboring CTs touched. However, while less than 1% of the nuclear volume represented domains of inter-chromosomal mixing, the dynamic plasticity of DNA foci within individual CTs allows continual transformation of CT structure so that different domains of chromatin mixing evolve over time. Notably, chromatin mixing at the boundaries of adjacent CTs had little impact on the innate structural properties of DNA foci. However, when TSA was used to alter the extent of histone acetylation changes in chromatin correlated with increased chromatin mixing. We propose that DNA foci maintain a structural integrity that restricts widespread mixing of DNA and discuss how the potential to dynamically remodel genome organization might alter during cell differentiation.


Chromosomes/genetics , Chromosomes/metabolism , DNA/genetics , DNA/metabolism , Cell Survival , Chromatin/genetics , Chromatin/metabolism , HeLa Cells , Humans , Staining and Labeling
17.
Trends Biochem Sci ; 36(8): 405-14, 2011 Aug.
Article En | MEDLINE | ID: mdl-21641805

Many replication origins that are licensed by loading MCM2-7 complexes in G1 are not normally used. Activation of these dormant origins during S phase provides a first line of defence for the genome if replication is inhibited. When replication forks fail, dormant origins are activated within regions of the genome currently engaged in replication. At the same time, DNA damage-response kinases activated by the stalled forks preferentially suppress the assembly of new replication factories, thereby ensuring that chromosomal regions experiencing replicative stress complete synthesis before new regions of the genome are replicated. Mice expressing reduced levels of MCM2-7 have fewer dormant origins, are cancer-prone and are genetically unstable, demonstrating the importance of dormant origins for preserving genome integrity. We review the function of dormant origins, the molecular mechanism of their regulation and their physiological implications.


Cell Cycle Proteins/genetics , Cell Cycle/genetics , DNA Replication/genetics , Nuclear Proteins/genetics , Replication Origin/genetics , Silencer Elements, Transcriptional/genetics , Animals , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Mice , Minichromosome Maintenance Complex Component 2 , Minichromosome Maintenance Complex Component 3 , Minichromosome Maintenance Complex Component 4 , Minichromosome Maintenance Complex Component 6 , Minichromosome Maintenance Complex Component 7 , S Phase/genetics , Stochastic Processes
18.
Curr Opin Genet Dev ; 20(6): 670-6, 2010 Dec.
Article En | MEDLINE | ID: mdl-20850963

The emergence of biological function from the dynamic control of cellular signalling molecules is a fundamental process in biology. Key questions include: How do cells decipher noisy environmental cues, encode these signals to control fate decisions and propagate information through tissues? Recent advances in systems biology, and molecular and cellular biology, exemplified by analyses of signalling via the transcription factor Nuclear Factor kappaB (NF-κB), reveal a critical role of oscillatory control in the regulation of these biological functions. The emerging view is that the oscillatory dynamics of signalling molecules and the epigenetically regulated specificity for target genes contribute to robust regulation of biological function at different levels of cellular organisation through frequency-dependent information encoding.


Biological Clocks , Signal Transduction , Animals , Epigenesis, Genetic , Humans , NF-kappa B/metabolism
19.
Essays Biochem ; 48(1): 25-43, 2010 Sep 20.
Article En | MEDLINE | ID: mdl-20822484

Eukaryotic cells are defined by the genetic information that is stored in their DNA. To function, this genetic information must be decoded. In doing this, the information encoded in DNA is copied first into RNA, during RNA transcription. Primary RNA transcripts are generated within transcription factories, where they are also processed into mature mRNAs, which then pass to the cytoplasm. In the cytoplasm these mRNAs can finally be translated into protein in order to express the genetic information as a functional product. With only rare exceptions, the cells of an individual multicellular eukaryote contain identical genetic information. However, as different genes must be expressed in different cell types to define the structure and function of individual tissues, it is clear that mechanisms must have evolved to regulate gene expression. In higher eukaryotes, mechanisms that regulate the interaction of DNA with the sites where nuclear functions are performed provide one such layer of regulation. In this chapter, I evaluate how a detailed understanding of nuclear structure and chromatin dynamics are beginning to reveal how spatial mechanisms link chromatin structure and function. As these mechanisms operate to modulate the genetic information in DNA, the regulation of chromatin function by nuclear architecture defines the concept of 'spatial epigenetics'.


Cell Nucleus/ultrastructure , Epigenomics , Cell Nucleus/metabolism , Chromatin/metabolism , Gene Expression , HeLa Cells , Humans , Microscopy, Electron
20.
Chromosome Res ; 18(5): 525-41, 2010 Jul.
Article En | MEDLINE | ID: mdl-20568006

Cell type and tissue architecture correlate with genome organization in higher eukaryotes, and structural nuclear landmarks are faithfully transmitted from one cell generation to the next. However, how nuclear components find their place in the nucleus after mitosis is still a matter of debate. As the major structural proteins within nuclei, the nuclear lamins are good candidates to re-establish nuclear compartments following mitosis. Human cells with reduced expression of the major B-type lamin protein, lamin B1, were generated using RNA interference. Mitotic and nuclear assembly phenotypes were then visualized in both fixed and living cells. Mitotic defects in lamin B1-depleted cells correlated with a general deterioration in nuclear compartmentalization and chromatin structure, frequent failure of chromosome segregation, and profound disorganization of centromeres. Examination of cells with normal lamin B1 expression indicated that small lamin B1 foci remain associated with major nuclear compartments--chromatin, nucleoli, and nuclear speckles--during an unperturbed mitosis. Our experiments show that normal lamin B1 expression is required for successful cell division and provide preliminary evidence that lamin B1-containing remnants of the interphase nucleoskeleton persist throughout mitosis. We suggest that these residual structures provide landmarks that are targeted during nuclear reassembly to allow key features of nuclear organization to be inherited from one cell cycle to the next.


Cell Nucleus/metabolism , Lamin Type B/physiology , Mitosis/physiology , Cell Nucleolus/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Lamin Type B/biosynthesis , Nuclear Proteins/metabolism , RNA Interference , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors
...