Instytut Podstawowych Problemów Techniki
Polskiej Akademii Nauk

Partnerzy

James Bagnall


Ostatnie publikacje
1.  Moran J., Feltham L., Bagnall J., Goldrick M., Lord E., Nettleton C., Spiller David G., Roberts I., Paszek P., Live-cell imaging reveals single-cell and population-level infection strategies of Listeria monocytogenes in macrophages, Frontiers in Immunology, ISSN: 1664-3224, DOI: 10.3389/fimmu.2023.1235675, Vol.14, pp.1235675-1-17, 2023

Streszczenie:
Pathogens have developed intricate strategies to overcome the host’s innate immune responses. In this paper we use live-cell microscopy with a single bacterium resolution to follow in real time interactions between the food-borne pathogen L. monocytogenes and host macrophages, a key event controlling the infection in vivo. We demonstrate that infection results in heterogeneous outcomes, with only a subset of bacteria able to establish a replicative invasion of macrophages. The fate of individual bacteria in the same host cell was independent from the host cell and non-cooperative, being independent from co-infecting bacteria. A higher multiplicity of infection resulted in a reduced probability of replication of the overall bacterial population. By use of internalisation assays and conditional probabilities to mathematically describe the two-stage invasion process, we demonstrate that the higher MOI compromises the ability of macrophages to phagocytose bacteria. We found that the rate of phagocytosis is mediated via the secreted Listeriolysin toxin (LLO), while the probability of replication of intracellular bacteria remained constant. Using strains expressing fluorescent reporters to follow transcription of either the LLO-encoding hly or actA genes, we show that replicative bacteria exhibited higher PrfA regulon expression in comparison to those bacteria that did not replicate, however elevated PrfA expression per se was not sufficient to increase the probability of replication. Overall, this demonstrates a new role for the population-level, but not single cell, PrfA-mediated activity to regulate outcomes of host pathogen interactions.

Słowa kluczowe:
Listeria monocytogenes, macrophage, single cell heterogeneity, phagocytosis, PrfA regulon, listeriolysin

Afiliacje autorów:
Moran J. - inna afiliacja
Feltham L. - inna afiliacja
Bagnall J. - inna afiliacja
Goldrick M. - inna afiliacja
Lord E. - inna afiliacja
Nettleton C. - inna afiliacja
Spiller David G. - inna afiliacja
Roberts I. - inna afiliacja
Paszek P. - IPPT PAN
140p.
2.  Downton P., Bagnall James S., England H., Spiller David G., Humphreys Neil E., Jackson Dean A., Paszek P., White Michael R.R., Adamson Antony D., Overexpression of IκB⍺ modulates NF-κB activation of inflammatory target gene expression, Frontiers in Molecular Biosciences, ISSN: 2296-889X, DOI: 10.3389/fmolb.2023.1187187, Vol.10, pp.1187187-1-15, 2023

Streszczenie:
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.

Słowa kluczowe:
NF-κB, inflammation, IκB⍺, overexpression, gene expression, localisation

Afiliacje autorów:
Downton P. - inna afiliacja
Bagnall James S. - inna afiliacja
England H. - inna afiliacja
Spiller David G. - inna afiliacja
Humphreys Neil E. - inna afiliacja
Jackson Dean A. - inna afiliacja
Paszek P. - IPPT PAN
White Michael R.R. - University of Manchester (GB)
Adamson Antony D. - inna afiliacja
140p.
3.  Kalliara E., Kardynska M., Bagnall J., Spiller David G., Müller W., Ruckerl D., Śmieja J., Biswas Subhra K., Paszek P., Post-transcriptional regulatory feedback encodes JAK-STAT signal memory of interferon stimulation, Frontiers in Immunology, ISSN: 1664-3224, DOI: 10.3389/fimmu.2022.947213, Vol.13, pp.947213-1-19, 2022

Streszczenie:
Immune cells fine tune their responses to infection and inflammatory cues. Here, using live-cell confocal microscopy and mathematical modelling, we investigate interferon-induced JAK-STAT signalling in innate immune macrophages. We demonstrate that transient exposure to IFN-γ stimulation induces a long-term desensitisation of STAT1 signalling and gene expression responses, revealing a dose- and time-dependent regulatory feedback that controls JAK-STAT responses upon re-exposure to stimulus. We show that IFN-α/β1 elicit different level of desensitisation from IFN-γ, where cells refractory to IFN-α/β1 are sensitive to IFN-γ, but not vice versa. We experimentally demonstrate that the underlying feedback mechanism involves regulation of STAT1 phosphorylation but is independent of new mRNA synthesis and cognate receptor expression. A new feedback model of the protein tyrosine phosphatase activity recapitulates experimental data and demonstrates JAK-STAT network’s ability to decode relative changes of dose, timing, and type of temporal interferon stimulation. These findings reveal that STAT desensitisation renders cells with signalling memory of type I and II interferon stimulation, which in the future may improve administration of interferon therapy.

Słowa kluczowe:
JAK-STAT network, STAT1 kinetics, interferons, pathway desensitisation, mathematical modelling, signal memory, live-cell microscopy

Afiliacje autorów:
Kalliara E. - inna afiliacja
Kardynska M. - inna afiliacja
Bagnall J. - inna afiliacja
Spiller David G. - inna afiliacja
Müller W. - inna afiliacja
Ruckerl D. - inna afiliacja
Śmieja J. - Silesian University of Technology (PL)
Biswas Subhra K. - inna afiliacja
Paszek P. - IPPT PAN
140p.
4.  Bagnall J., Rowe W., Alachkar N., Roberts J., England H., Clark C., Platt M., Jackson Dean A., Muldoon M., Paszek P., Gene-Specific Linear Trends Constrain Transcriptional Variability of the Toll-like Receptor Signaling, Cell Systems, ISSN: 2405-4712, DOI: 10.1016/j.cels.2020.08.007, Vol.11, No.3, pp.300-314, 2020

Streszczenie:
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-1β 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.

Słowa kluczowe:
cellular heterogeneity, transcriptional bursting, stochastic gene expression, toll-like receptor, single-cell transcriptomics, stochastic modeling, TNF-α, IL-1β

Afiliacje autorów:
Bagnall J. - inna afiliacja
Rowe W. - inna afiliacja
Alachkar N. - inna afiliacja
Roberts J. - inna afiliacja
England H. - inna afiliacja
Clark C. - inna afiliacja
Platt M. - inna afiliacja
Jackson Dean A. - inna afiliacja
Muldoon M. - inna afiliacja
Paszek P. - inna afiliacja
200p.
5.  Paszek A., Kardyńska M., Bagnall J., Śmieja J., Spiller David G., Widłak P., Kimmel M., Wiesława W., Paszek P., Heat shock response regulates stimulus-specificity and sensitivity of the pro-inflammatory NF-κB signalling, Cell Communication and Signaling, ISSN: 1478-811X, DOI: 10.1186/s12964-020-00583-0, Vol.18, pp.77-1-21, 2020

Streszczenie:
Background

Ability to adapt to temperature changes trough the Heat Shock Response (HSR) pathways is one of the most fundamental and clinically relevant cellular response systems. Heat Shock (HS) affects the signalling and gene expression responses of the Nuclear Factor κB (NF-κB) transcription factor, a critical regulator of proliferation and inflammation, however, our quantitative understanding of how cells sense and adapt to temperature changes is limited.
Methods

We used live-cell time-lapse microscopy and mathematical modelling to understand the signalling of the NF-κB system in the human MCF7 breast adenocarcinoma cells in response to pro-inflammatory Interleukin 1β (IL1β) and Tumour Necrosis Factor α (TNFα) cytokines, following exposure to a 37–43 °C range of physiological and clinical temperatures.
Results

We show that exposure to 43 °C 1 h HS inhibits the immediate NF-κB signalling response to TNFα and IL1β stimulation although uptake of cytokines is not impaired. Within 4 h after HS treatment IL1β-induced NF-κB responses return to normal levels, but the recovery of the TNFα-induced responses is still affected. Using siRNA knock-down of Heat Shock Factor 1 (HSF1) we show that this stimulus-specificity is conferred via the Inhibitory κB kinase (IKK) signalosome where HSF1-dependent feedback regulates TNFα, but not IL1β-mediated IKK recovery post HS. Furthermore, we demonstrate that through the temperature-dependent denaturation and recovery of IKK, TNFα and IL1β-mediated signalling exhibit different temperature sensitivity and adaptation to repeated HS when exposed to a 37–43 °C temperature range. Specifically, IL1β-mediated NF-κB responses are more robust to temperature changes in comparison to those induced by TNFα treatment.

Afiliacje autorów:
Paszek A. - inna afiliacja
Kardyńska M. - inna afiliacja
Bagnall J. - inna afiliacja
Śmieja J. - Silesian University of Technology (PL)
Spiller David G. - inna afiliacja
Widłak P. - inna afiliacja
Kimmel M. - Rice University (US)
Wiesława W. - inna afiliacja
Paszek P. - IPPT PAN
6.  Bagnall J., Boddington C., England H., Brignall R., Downton P., Alsoufi Z., Boyd J., Rowe W., Bennett A., Walker C., Adamson A., Patel Nisha M. X., O’Cualain R., Schmidt L., Spiller David G., Jackson Dean A., Müller W., Muldoon M., White Michael R. H.R., Paszek P., Quantitative analysis of competitive cytokine signaling predicts tissue thresholds for the propagation of macrophage activation, Science Signaling, ISSN: 1945-0877, DOI: 10.1126/scisignal.aaf3998, Vol.11, No.540, pp.1-15, 2018

Streszczenie:
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.

Afiliacje autorów:
Bagnall J. - inna afiliacja
Boddington C. - inna afiliacja
England H. - inna afiliacja
Brignall R. - inna afiliacja
Downton P. - inna afiliacja
Alsoufi Z. - inna afiliacja
Boyd J. - inna afiliacja
Rowe W. - inna afiliacja
Bennett A. - inna afiliacja
Walker C. - inna afiliacja
Adamson A. - inna afiliacja
Patel Nisha M. X. - inna afiliacja
O’Cualain R. - inna afiliacja
Schmidt L. - inna afiliacja
Spiller David G. - inna afiliacja
Jackson Dean A. - inna afiliacja
Müller W. - inna afiliacja
Muldoon M. - inna afiliacja
White Michael R. H.R. - University of Manchester (GB)
Paszek P. - inna afiliacja
40p.
7.  Brignall R., Cauchy P., Bevington Sarah L., Gorman B., Pisco Angela O., Bagnall J., Boddington C., Rowe W., England H., Rich K., Schmidt L., Dyer Nigel P., Travis Mark A., Ott S., Jackson Dean A., Cockerill Peter N., Paszek P., Integration of Kinase and Calcium Signaling at the Level of Chromatin Underlies Inducible Gene Activation in T Cells, JOURNAL OF IMMUNOLOGY, ISSN: 0022-1767, DOI: 10.4049/jimmunol.1602033, Vol.199, No.8, pp.2652-2667, 2017

Streszczenie:
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.

Afiliacje autorów:
Brignall R. - inna afiliacja
Cauchy P. - inna afiliacja
Bevington Sarah L. - inna afiliacja
Gorman B. - inna afiliacja
Pisco Angela O. - inna afiliacja
Bagnall J. - inna afiliacja
Boddington C. - inna afiliacja
Rowe W. - inna afiliacja
England H. - inna afiliacja
Rich K. - inna afiliacja
Schmidt L. - inna afiliacja
Dyer Nigel P. - inna afiliacja
Travis Mark A. - inna afiliacja
Ott S. - inna afiliacja
Jackson Dean A. - inna afiliacja
Cockerill Peter N. - inna afiliacja
Paszek P. - inna afiliacja
8.  Daniels Michael J.D., Rivers-Auty J., Schilling T., Spencer Nicholas G., Watremez W., Fasolino V., Booth Sophie J., White Claire S., Baldwin Alex G., Freeman S., Wong R., Latta C., Yu S., Jackson J., Fischer N., Koziel V., Pillot T., Bagnall J., Allan Stuart M., Paszek P., Galea J., Harte Michael K., Eder C., Lawrence Catherine B., Brough D., Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models, Nature Communications, ISSN: 2041-1723, DOI: 10.1038/ncomms12504, Vol.7, pp.12504-1-10, 2016

Streszczenie:
Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit cyclooxygenase-1 (COX-1) and COX-2 enzymes. The NLRP3 inflammasome is a multi-protein complex responsible for the processing of the proinflammatory cytokine interleukin-1β and is implicated in many inflammatory diseases. Here we show that several clinically approved and widely used NSAIDs of the fenamate class are effective and selective inhibitors of the NLRP3 inflammasome via inhibition of the volume-regulated anion channel in macrophages, independently of COX enzymes. Flufenamic acid and mefenamic acid are efficacious in NLRP3-dependent rodent models of inflammation in air pouch and peritoneum. We also show therapeutic effects of fenamates using a model of amyloid beta induced memory loss and a transgenic mouse model of Alzheimer’s disease. These data suggest that fenamate NSAIDs could be repurposed as NLRP3 inflammasome inhibitors and Alzheimer’s disease therapeutics.

Afiliacje autorów:
Daniels Michael J.D. - inna afiliacja
Rivers-Auty J. - inna afiliacja
Schilling T. - inna afiliacja
Spencer Nicholas G. - inna afiliacja
Watremez W. - inna afiliacja
Fasolino V. - inna afiliacja
Booth Sophie J. - inna afiliacja
White Claire S. - inna afiliacja
Baldwin Alex G. - inna afiliacja
Freeman S. - inna afiliacja
Wong R. - inna afiliacja
Latta C. - inna afiliacja
Yu S. - inna afiliacja
Jackson J. - inna afiliacja
Fischer N. - inna afiliacja
Koziel V. - inna afiliacja
Pillot T. - inna afiliacja
Bagnall J. - inna afiliacja
Allan Stuart M. - inna afiliacja
Paszek P. - inna afiliacja
Galea J. - inna afiliacja
Harte Michael K. - inna afiliacja
Eder C. - inna afiliacja
Lawrence Catherine B. - inna afiliacja
Brough D. - inna afiliacja
45p.
9.  Adamson A., Boddington C., Downton P., Rowe W., Bagnall J., Lam C., Maya-Mendoza A., Schmidt L., Harper Claire V.V., Spiller David G., Rand David A.A., Jackson Dean A., White Michael R. H.R., Paszek P., Signal transduction controls heterogeneous NF-κB dynamics and target gene expression through cytokine-specific refractory states, Nature Communications, ISSN: 2041-1723, DOI: 10.1038/ncomms12057, Vol.7, pp.12057-1-14, 2016

Streszczenie:
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-1β 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.

Afiliacje autorów:
Adamson A. - inna afiliacja
Boddington C. - inna afiliacja
Downton P. - inna afiliacja
Rowe W. - inna afiliacja
Bagnall J. - inna afiliacja
Lam C. - inna afiliacja
Maya-Mendoza A. - inna afiliacja
Schmidt L. - inna afiliacja
Harper Claire V.V. - University of Manchester (GB)
Spiller David G. - inna afiliacja
Rand David A.A. - University of Warwick (GB)
Jackson Dean A. - inna afiliacja
White Michael R. H.R. - University of Manchester (GB)
Paszek P. - inna afiliacja
45p.
10.  Phillips N., Manning C., Pettini T., Veronica B., Elli M., Peter S., Boyd J., Bagnall J., Paszek P., Spiller David G., White M., Goodfellow M., Tobias G., Magnus R., Nancy P., Stochasticity in the miR-9/Hes1 oscillatory network can account for clonal heterogeneity in the timing of differentiation, eLife, ISSN: 2050-084X, DOI: 10.7554/eLife.16118, Vol.5, pp.e16118-1-33, 2016

Streszczenie:
Recent studies suggest that cells make stochastic choices with respect to differentiation or division. However, the molecular mechanism underlying such stochasticity is unknown. We previously proposed that the timing of vertebrate neuronal differentiation is regulated by molecular oscillations of a transcriptional repressor, HES1, tuned by a post-transcriptional repressor, miR-9. Here, we computationally model the effects of intrinsic noise on the Hes1/miR-9 oscillator as a consequence of low molecular numbers of interacting species, determined experimentally. We report that increased stochasticity spreads the timing of differentiation in a population, such that initially equivalent cells differentiate over a period of time. Surprisingly, inherent stochasticity also increases the robustness of the progenitor state and lessens the impact of unequal, random distribution of molecules at cell division on the temporal spread of differentiation at the population level. This advantageous use of biological noise contrasts with the view that noise needs to be counteracted.

Afiliacje autorów:
Phillips N. - inna afiliacja
Manning C. - inna afiliacja
Pettini T. - inna afiliacja
Veronica B. - inna afiliacja
Elli M. - inna afiliacja
Peter S. - inna afiliacja
Boyd J. - inna afiliacja
Bagnall J. - inna afiliacja
Paszek P. - IPPT PAN
Spiller David G. - inna afiliacja
White M. - inna afiliacja
Goodfellow M. - inna afiliacja
Tobias G. - inna afiliacja
Magnus R. - inna afiliacja
Nancy P. - inna afiliacja
11.  Martín-Sánchez F., Diamond C., Zeitler M., Gomez A., Baroja-Mazo A., Bagnall J., Spiller David G., White M.R., Daniels Michael J.D., Mortellaro A., Peñalver M., Paszek P., Steringer J., Nickel W., Brough D., Pelegrín P., Inflammasome-dependent IL-1β release depends upon membrane permeabilisation, Cell Death & Differentiation, ISSN: 1350-9047, DOI: 10.1038/cdd.2015.176, Vol.23, pp.1219-1231, 2016

Streszczenie:
Interleukin-1β (IL-1β) is a critical regulator of the inflammatory response. IL-1β is not secreted through the conventional ER–Golgi route of protein secretion, and to date its mechanism of release has been unknown. Crucially, its secretion depends upon the processing of a precursor form following the activation of the multimolecular inflammasome complex. Using a novel and reversible pharmacological inhibitor of the IL-1β release process, in combination with biochemical, biophysical, and real-time single-cell confocal microscopy with macrophage cells expressing Venus-labelled IL-1β, we have discovered that the secretion of IL-1β after inflammasome activation requires membrane permeabilisation, and occurs in parallel with the death of the secreting cell. Thus, in macrophages the release of IL-1β in response to inflammasome activation appears to be a secretory process independent of nonspecific leakage of proteins during cell death. The mechanism of membrane permeabilisation leading to IL-1β release is distinct from the unconventional secretory mechanism employed by its structural homologues fibroblast growth factor 2 (FGF2) or IL-1α, a process that involves the formation of membrane pores but does not result in cell death. These discoveries reveal key processes at the initiation of an inflammatory response and deliver new insights into the mechanisms of protein release.

Afiliacje autorów:
Martín-Sánchez F. - inna afiliacja
Diamond C. - inna afiliacja
Zeitler M. - inna afiliacja
Gomez A. - inna afiliacja
Baroja-Mazo A. - inna afiliacja
Bagnall J. - inna afiliacja
Spiller David G. - inna afiliacja
White M.R. - University of Manchester (GB)
Daniels Michael J.D. - inna afiliacja
Mortellaro A. - inna afiliacja
Peñalver M. - inna afiliacja
Paszek P. - inna afiliacja
Steringer J. - inna afiliacja
Nickel W. - inna afiliacja
Brough D. - inna afiliacja
Pelegrín P. - inna afiliacja

Kategoria A Plus

IPPT PAN

logo ippt            ul. Pawińskiego 5B, 02-106 Warszawa
  +48 22 826 12 81 (centrala)
  +48 22 826 98 15
 

Znajdź nas

mapka
© Instytut Podstawowych Problemów Techniki Polskiej Akademii Nauk 2024