Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
EMBO Mol Med ; 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750308

RÉSUMÉ

Necroptosis is a lytic form of regulated cell death reported to contribute to inflammatory diseases of the gut, skin and lung, as well as ischemic-reperfusion injuries of the kidney, heart and brain. However, precise identification of the cells and tissues that undergo necroptotic cell death in vivo has proven challenging in the absence of robust protocols for immunohistochemical detection. Here, we provide automated immunohistochemistry protocols to detect core necroptosis regulators - Caspase-8, RIPK1, RIPK3 and MLKL - in formalin-fixed mouse and human tissues. We observed surprising heterogeneity in protein expression within tissues, whereby short-lived immune barrier cells were replete with necroptotic effectors, whereas long-lived cells lacked RIPK3 or MLKL expression. Local changes in the expression of necroptotic effectors occurred in response to insults such as inflammation, dysbiosis or immune challenge, consistent with necroptosis being dysregulated in disease contexts. These methods will facilitate the precise localisation and evaluation of necroptotic signaling in vivo.

2.
Nat Commun ; 14(1): 6046, 2023 09 28.
Article de Anglais | MEDLINE | ID: mdl-37770424

RÉSUMÉ

Across the globe, 2-3% of humans carry the p.Ser132Pro single nucleotide polymorphism in MLKL, the terminal effector protein of the inflammatory form of programmed cell death, necroptosis. Here we show that this substitution confers a gain in necroptotic function in human cells, with more rapid accumulation of activated MLKLS132P in biological membranes and MLKLS132P overriding pharmacological and endogenous inhibition of MLKL. In mouse cells, the equivalent Mlkl S131P mutation confers a gene dosage dependent reduction in sensitivity to TNF-induced necroptosis in both hematopoietic and non-hematopoietic cells, but enhanced sensitivity to IFN-ß induced death in non-hematopoietic cells. In vivo, MlklS131P homozygosity reduces the capacity to clear Salmonella from major organs and retards recovery of hematopoietic stem cells. Thus, by dysregulating necroptosis, the S131P substitution impairs the return to homeostasis after systemic challenge. Present day carriers of the MLKL S132P polymorphism may be the key to understanding how MLKL and necroptosis modulate the progression of complex polygenic human disease.


Sujet(s)
Apoptose , Protein kinases , Humains , Animaux , Souris , Phosphorylation , Protein kinases/génétique , Protein kinases/métabolisme , Membrane cellulaire/métabolisme , Mutation , Facteurs de transcription/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme
3.
Biochem J ; 480(9): 665-684, 2023 05 15.
Article de Anglais | MEDLINE | ID: mdl-37115711

RÉSUMÉ

Necroptosis is a mode of programmed, lytic cell death that is executed by the mixed lineage kinase domain-like (MLKL) pseudokinase following activation by the upstream kinases, receptor-interacting serine/threonine protein kinase (RIPK)-1 and RIPK3. Dysregulated necroptosis has been implicated in the pathophysiology of many human diseases, including inflammatory and degenerative conditions, infectious diseases and cancers, provoking interest in pharmacological targeting of the pathway. To identify small molecules impacting on the necroptotic machinery, we performed a phenotypic screen using a mouse cell line expressing an MLKL mutant that kills cells in the absence of upstream death or pathogen detector receptor activation. This screen identified the vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR) tyrosine kinase inhibitor, ABT-869 (Linifanib), as a small molecule inhibitor of necroptosis. We applied a suite of cellular, biochemical and biophysical analyses to pinpoint the apical necroptotic kinase, RIPK1, as the target of ABT-869 inhibition. Our study adds to the repertoire of established protein kinase inhibitors that additionally target RIPK1 and raises the prospect that serendipitous targeting of necroptosis signalling may contribute to their clinical efficacy in some settings.


Sujet(s)
Protein kinases , Humains , Protein kinases/génétique , Protein kinases/métabolisme , Nécroptose , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Apoptose , Récepteurs aux facteurs de croissance endothéliale vasculaire/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme
4.
Methods Enzymol ; 667: 229-273, 2022.
Article de Anglais | MEDLINE | ID: mdl-35525543

RÉSUMÉ

The non-catalytic cousins of protein kinases, the pseudokinases, have grown to prominence as indispensable signaling entities over the past decade, despite their lack of catalytic activity. Because their importance has only been fully embraced recently, many of the 10% of the human kinome categorized as pseudokinases are yet to be attributed biological functions. The advent of CRISPR-Cas9 editing to genetically delete pseudokinases in a cell line of interest has proven invaluable to dissecting many functions and remains the method of choice for gene knockout. Here, using the terminal effector pseudokinase in the necroptosis cell death pathway, MLKL, as an exemplar, we describe a method for genetic knockout of pseudokinases in cultured cells. This method does not retain the CRISPR guide sequence in the edited cells, which eliminates possible interference in subsequent reconstitution studies where mutant forms of the pseudokinase can be reintroduced into cells exogenously for detailed mechanistic characterization.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Protein kinases , Lignée cellulaire , Édition de gène/méthodes , Humains , Protein kinases/génétique
5.
Nat Commun ; 13(1): 2073, 2022 04 19.
Article de Anglais | MEDLINE | ID: mdl-35440107

RÉSUMÉ

Modulation of protein abundance using tag-Targeted Protein Degrader (tTPD) systems targeting FKBP12F36V (dTAGs) or HaloTag7 (HaloPROTACs) are powerful approaches for preclinical target validation. Interchanging tags and tag-targeting degraders is important to achieve efficient substrate degradation, yet limited degrader/tag pairs are available and side-by-side comparisons have not been performed. To expand the tTPD repertoire we developed catalytic NanoLuc-targeting PROTACs (NanoTACs) to hijack the CRL4CRBN complex and degrade NanoLuc tagged substrates, enabling rapid luminescence-based degradation screening. To benchmark NanoTACs against existing tTPD systems we use an interchangeable reporter system to comparatively test optimal degrader/tag pairs. Overall, we find the dTAG system exhibits superior degradation. To align tag-induced degradation with physiology we demonstrate that NanoTACs limit MLKL-driven necroptosis. In this work we extend the tTPD platform to include NanoTACs adding flexibility to tTPD studies, and benchmark each tTPD system to highlight the importance of comparing each system against each substrate.


Sujet(s)
Référenciation , Protéine 1A de liaison au tacrolimus , Luciferases , Protéolyse , Protéine 1A de liaison au tacrolimus/génétique
6.
Cell Death Dis ; 13(4): 291, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35365636

RÉSUMÉ

Necroptosis is a form of caspase-independent programmed cell death that arises from disruption of cell membranes by the mixed lineage kinase domain-like (MLKL) pseudokinase after its activation by the upstream kinases, receptor interacting protein kinase (RIPK)-1 and RIPK3, within a complex known as the necrosome. Dysregulated necroptosis has been implicated in numerous inflammatory pathologies. As such, new small molecule necroptosis inhibitors are of great interest, particularly ones that operate downstream of MLKL activation, where the pathway is less well defined. To better understand the mechanisms involved in necroptosis downstream of MLKL activation, and potentially uncover new targets for inhibition, we screened known kinase inhibitors against an activated mouse MLKL mutant, leading us to identify the lymphocyte-specific protein tyrosine kinase (Lck) inhibitor AMG-47a as an inhibitor of necroptosis. We show that AMG-47a interacts with both RIPK1 and RIPK3, that its ability to protect from cell death is dependent on the strength of the necroptotic stimulus, and that it blocks necroptosis most effectively in human cells. Moreover, in human cell lines, we demonstrate that AMG-47a can protect against cell death caused by forced dimerisation of MLKL truncation mutants in the absence of any upstream signalling, validating that it targets a process downstream of MLKL activation. Surprisingly, however, we also found that the cell death driven by activated MLKL in this model was completely dependent on the presence of RIPK1, and to a lesser extent RIPK3, although it was not affected by known inhibitors of these kinases. Together, these results suggest an additional role for RIPK1, or the necrosome, in mediating human necroptosis after MLKL is phosphorylated by RIPK3 and provide further insight into reported differences in the progression of necroptosis between mouse and human cells.


Sujet(s)
Nécroptose , Protein kinases , Animaux , Apoptose , Mort cellulaire , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes , Souris , Protein kinases/génétique , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Transduction du signal
7.
Cell Death Differ ; 29(9): 1804-1815, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35264780

RÉSUMÉ

Necroptosis is a lytic programmed cell death pathway with origins in innate immunity that is frequently dysregulated in inflammatory diseases. The terminal effector of the pathway, MLKL, is licensed to kill following phosphorylation of its pseudokinase domain by the upstream regulator, RIPK3 kinase. Phosphorylation provokes the unleashing of MLKL's N-terminal four-helix bundle (4HB or HeLo) domain, which binds and permeabilizes the plasma membrane to cause cell death. The precise mechanism by which the 4HB domain permeabilizes membranes, and how the mechanism differs between species, remains unclear. Here, we identify the membrane binding epitope of mouse MLKL using NMR spectroscopy. Using liposome permeabilization and cell death assays, we validate K69 in the α3 helix, W108 in the α4 helix, and R137/Q138 in the first brace helix as crucial residues for necroptotic signaling. This epitope differs from the phospholipid binding site reported for human MLKL, which comprises basic residues primarily located in the α1 and α2 helices. In further contrast to human and plant MLKL orthologs, in which the α3-α4 loop forms a helix, this loop is unstructured in mouse MLKL in solution. Together, these findings illustrate the versatility of the 4HB domain fold, whose lytic function can be mediated by distinct epitopes in different orthologs.


Sujet(s)
Nécroptose , Protein kinases , Animaux , Épitopes , Humains , Souris , Nécrose , Phosphorylation , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme
8.
Nat Commun ; 12(1): 2211, 2021 04 13.
Article de Anglais | MEDLINE | ID: mdl-33850121

RÉSUMÉ

Phosphorylation of the MLKL pseudokinase by the RIPK3 kinase leads to MLKL oligomerization, translocation to, and permeabilization of, the plasma membrane to induce necroptotic cell death. The precise choreography of MLKL activation remains incompletely understood. Here, we report Monobodies, synthetic binding proteins, that bind the pseudokinase domain of MLKL within human cells and their crystal structures in complex with the human MLKL pseudokinase domain. While Monobody-32 constitutively binds the MLKL hinge region, Monobody-27 binds MLKL via an epitope that overlaps the RIPK3 binding site and is only exposed after phosphorylated MLKL disengages from RIPK3 following necroptotic stimulation. The crystal structures identified two distinct conformations of the MLKL pseudokinase domain, supporting the idea that a conformational transition accompanies MLKL disengagement from RIPK3. These studies provide further evidence that MLKL undergoes a large conformational change upon activation, and identify MLKL disengagement from RIPK3 as a key regulatory step in the necroptosis pathway.


Sujet(s)
Mort cellulaire/physiologie , Nécroptose/physiologie , Protein kinases/composition chimique , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/composition chimique , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Animaux , Sites de fixation , Membrane cellulaire , Cristallographie aux rayons X , Cellules HT29 , Humains , Souris , Conformation moléculaire , Simulation de dynamique moléculaire , Mutation , Phosphorylation , Conformation des protéines , Protein kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Protéines recombinantes , Alignement de séquences , Cellules U937
9.
Cell Death Differ ; 28(7): 2126-2144, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33589776

RÉSUMÉ

Necroptosis is a lytic, inflammatory cell death pathway that is dysregulated in many human pathologies. The pathway is executed by a core machinery comprising the RIPK1 and RIPK3 kinases, which assemble into necrosomes in the cytoplasm, and the terminal effector pseudokinase, MLKL. RIPK3-mediated phosphorylation of MLKL induces oligomerization and translocation to the plasma membrane where MLKL accumulates as hotspots and perturbs the lipid bilayer to cause death. The precise choreography of events in the pathway, where they occur within cells, and pathway differences between species, are of immense interest. However, they have been poorly characterized due to a dearth of validated antibodies for microscopy studies. Here, we describe a toolbox of antibodies for immunofluorescent detection of the core necroptosis effectors, RIPK1, RIPK3, and MLKL, and their phosphorylated forms, in human and mouse cells. By comparing reactivity with endogenous proteins in wild-type cells and knockout controls in basal and necroptosis-inducing conditions, we characterise the specificity of frequently-used commercial and recently-developed antibodies for detection of necroptosis signaling events. Importantly, our findings demonstrate that not all frequently-used antibodies are suitable for monitoring necroptosis by immunofluorescence microscopy, and methanol- is preferable to paraformaldehyde-fixation for robust detection of specific RIPK1, RIPK3, and MLKL signals.


Sujet(s)
Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Animaux , Membrane cellulaire/métabolisme , Cellules HT29 , Humains , Souris , Nécroptose , Phosphorylation
10.
ACS Chem Biol ; 15(10): 2702-2713, 2020 10 16.
Article de Anglais | MEDLINE | ID: mdl-32902249

RÉSUMÉ

Necroptosis is an inflammatory form of programmed cell death that has been implicated in various human diseases. Compound 2 is a more potent analogue of the published compound 1 and inhibits necroptosis in human and murine cells at nanomolar concentrations. Several target engagement strategies were employed, including cellular thermal shift assays (CETSA) and diazirine-mediated photoaffinity labeling via a bifunctional photoaffinity probe derived from compound 2. These target engagement studies demonstrate that compound 2 binds to all three necroptotic effector proteins (mixed lineage kinase domain-like protein (MLKL), receptor-interacting serine/threonine protein kinase 1 (RIPK1) and receptor-interacting serine/threonine protein kinase 3 (RIPK3)) at different levels in vitro and in cells. Compound 2 also shows efficacy in vivo in a murine model of systemic inflammatory response syndrome (SIRS).


Sujet(s)
Nécroptose/effets des médicaments et des substances chimiques , Phénylurées/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Sulfonamides/usage thérapeutique , Animaux , Lignée cellulaire tumorale , Femelle , Humains , Souris de lignée C57BL , Phénylurées/métabolisme , Phénylurées/pharmacocinétique , Liaison aux protéines , Inhibiteurs de protéines kinases/métabolisme , Inhibiteurs de protéines kinases/pharmacocinétique , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/antagonistes et inhibiteurs , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Sulfonamides/métabolisme , Sulfonamides/pharmacocinétique , Syndrome de réponse inflammatoire généralisée/traitement médicamenteux
11.
Nat Commun ; 11(1): 3151, 2020 06 19.
Article de Anglais | MEDLINE | ID: mdl-32561730

RÉSUMÉ

Mixed lineage kinase domain-like (MLKL) is the terminal protein in the pro-inflammatory necroptotic cell death program. RIPK3-mediated phosphorylation is thought to initiate MLKL oligomerization, membrane translocation and membrane disruption, although the precise choreography of events is incompletely understood. Here, we use single-cell imaging approaches to map the chronology of endogenous human MLKL activation during necroptosis. During the effector phase of necroptosis, we observe that phosphorylated MLKL assembles into higher order species on presumed cytoplasmic necrosomes. Subsequently, MLKL co-traffics with tight junction proteins to the cell periphery via Golgi-microtubule-actin-dependent mechanisms. MLKL and tight junction proteins then steadily co-accumulate at the plasma membrane as heterogeneous micron-sized hotspots. Our studies identify MLKL trafficking and plasma membrane accumulation as crucial necroptosis checkpoints. Furthermore, the accumulation of phosphorylated MLKL at intercellular junctions accelerates necroptosis between neighbouring cells, which may be relevant to inflammatory bowel disease and other necroptosis-mediated enteropathies.


Sujet(s)
Nécroptose , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Animaux , Lignée cellulaire , Membrane cellulaire/métabolisme , Humains , Transport des protéines , Protéines de la jonction serrée/métabolisme
12.
Cell Rep ; 28(13): 3309-3319.e5, 2019 09 24.
Article de Anglais | MEDLINE | ID: mdl-31553902

RÉSUMÉ

Necroptotic cell death has been implicated in many human pathologies and is thought to have evolved as an innate immunity mechanism. The pathway relies on two key effectors: the kinase receptor-interacting protein kinase 3 (RIPK3) and the terminal effector, the pseudokinase mixed-lineage kinase-domain-like (MLKL). We identify proteins with high sequence similarity to the pseudokinase domain of MLKL in poxvirus genomes. Expression of these proteins from the BeAn 58058 and Cotia poxviruses, but not swinepox, in human and mouse cells blocks cellular MLKL activation and necroptotic cell death. We show that viral MLKL-like proteins function as dominant-negative mimics of host MLKL, which inhibit necroptosis by sequestering RIPK3 via its kinase domain to thwart MLKL engagement and phosphorylation. These data support an ancestral role for necroptosis in defense against pathogens. Furthermore, mimicry of a cellular pseudokinase by a pathogen adds to the growing repertoire of functions performed by pseudokinases in signal transduction.


Sujet(s)
Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Animaux , Mort cellulaire , Humains , Immunité innée , Souris , Nécrose
13.
Cell Death Differ ; 26(9): 1631-1645, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-30442947

RÉSUMÉ

Different forms of regulated cell death-like apoptosis and necroptosis contribute to the pathophysiology of clinical conditions including ischemia-reperfusion injury, myocardial infarction, sepsis, and multiple sclerosis. In particular, the kinase activity of the receptor-interacting serine/threonine protein kinase 1 (RIPK1) is crucial for cell fate in inflammation and cell death. However, despite its involvement in pathological conditions, no pharmacologic inhibitor of RIPK1-mediated cell death is currently in clinical use. Herein, we screened a collection of clinical compounds to assess their ability to modulate RIPK1-mediated cell death. Our small-scale screen identified the anti-epilepsy drug Phenhydan® as a potent inhibitor of death receptor-induced necroptosis and apoptosis. Accordingly, Phenhydan® blocked activation of necrosome formation/activation as well as death receptor-induced NF-κB signaling by influencing the membrane function of cells, such as lipid raft formation, thus exerting an inhibitory effect on pathophysiologic cell death processes. By targeting death receptor signaling, the already FDA-approved Phenhydan® may provide new therapeutic strategies for inflammation-driven diseases caused by aberrant cell death.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Nécroptose/effets des médicaments et des substances chimiques , Phénytoïne/pharmacologie , Animaux , Anticonvulsivants/pharmacologie , Apoptose/génétique , Mort cellulaire/effets des médicaments et des substances chimiques , Mort cellulaire/génétique , Cellules HT29 , Humains , Inflammation/génétique , Inflammation/anatomopathologie , Souris , Sclérose en plaques/traitement médicamenteux , Sclérose en plaques/génétique , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/génétique , Cellules NIH 3T3 , Nécroptose/génétique , Phénytoïne/usage thérapeutique , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Récepteurs à domaine de mort/antagonistes et inhibiteurs , Récepteurs à domaine de mort/génétique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/génétique , Sepsie/traitement médicamenteux , Sepsie/génétique
14.
Nat Commun ; 9(1): 2422, 2018 06 21.
Article de Anglais | MEDLINE | ID: mdl-29930286

RÉSUMÉ

Necroptotic cell death is mediated by the most terminal known effector of the pathway, MLKL. Precisely how phosphorylation of the MLKL pseudokinase domain activation loop by the upstream kinase, RIPK3, induces unmasking of the N-terminal executioner four-helix bundle (4HB) domain of MLKL, higher-order assemblies, and permeabilization of plasma membranes remains poorly understood. Here, we reveal the existence of a basal monomeric MLKL conformer present in human cells prior to exposure to a necroptotic stimulus. Following activation, toggling within the MLKL pseudokinase domain promotes 4HB domain disengagement from the pseudokinase domain αC helix and pseudocatalytic loop, to enable formation of a necroptosis-inducing tetramer. In contrast to mouse MLKL, substitution of RIPK3 substrate sites in the human MLKL pseudokinase domain completely abrogated necroptotic signaling. Therefore, while the pseudokinase domains of mouse and human MLKL function as molecular switches to control MLKL activation, the underlying mechanism differs between species.


Sujet(s)
Mort cellulaire/physiologie , Modèles moléculaires , Protein kinases/physiologie , Animaux , Bases de données de protéines , Humains , Spectrométrie de masse , Souris , Polymérisation , Conformation des protéines , Domaines protéiques , Protein kinases/composition chimique , Protein kinases/génétique , Transduction du signal , Spécificité d'espèce
15.
Environ Sci Pollut Res Int ; 25(23): 23074-23081, 2018 Aug.
Article de Anglais | MEDLINE | ID: mdl-29860686

RÉSUMÉ

Per- and polyfluoroalkyl substances (PFAS) are synthetic surfactants with a wide variety of applications; however, due to their stability, they are particularly resistant to degradation and, as such, are classed as persistent organic pollutants. Perfluorooctane sulfonate (PFOS) is one such PFAS that is still detectable in a range of different environmental settings, despite its use now being regulated in numerous countries. Elevated levels of PFOS have been detected in various avian species, and the impact of this on avian health is of interest when determining acceptable levels of PFOS in the environment. Due to its similarities to naturally occurring fatty acids, PFOS has potential to disrupt a range of biological pathways, particularly those associated with lipid metabolism, and this has been shown in various species. In this study, we have investigated how in ovo exposure to environmentally relevant levels of PFOS affects expression of genes involved in lipid metabolism of developing chicken embryos. We have found a broad suppression of transcription of genes involved in fatty acid oxidation and PPAR-mediated transcription with more significant effects apparent at lower doses of PFOS. These results highlight the need for more research investigating the biological impacts of low levels of PFAS to properly inform environmental policy governing their regulation.


Sujet(s)
Acides alcanesulfoniques/effets indésirables , Polluants environnementaux/effets indésirables , Acides gras/métabolisme , Fluorocarbones/effets indésirables , Expression des gènes/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Animaux , Embryon de poulet , Relation dose-effet des médicaments , Réaction de polymérisation en chaîne
16.
Biochem Soc Trans ; 45(3): 665-681, 2017 06 15.
Article de Anglais | MEDLINE | ID: mdl-28620028

RÉSUMÉ

Over the past decade, our understanding of the mechanisms by which pseudokinases, which comprise ∼10% of the human and mouse kinomes, mediate signal transduction has advanced rapidly with increasing structural, biochemical, cellular and genetic studies. Pseudokinases are the catalytically defective counterparts of conventional, active protein kinases and have been attributed functions as protein interaction domains acting variously as allosteric modulators of conventional protein kinases and other enzymes, as regulators of protein trafficking or localisation, as hubs to nucleate assembly of signalling complexes, and as transmembrane effectors of such functions. Here, by categorising mammalian pseudokinases based on their known functions, we illustrate the mechanistic diversity among these proteins, which can be viewed as a window into understanding the non-catalytic functions that can be exerted by conventional protein kinases.


Sujet(s)
Protein kinases/métabolisme , Transduction du signal , Régulation allostérique , Animaux , Humains , Protéines membranaires/métabolisme , Protein kinases/physiologie , Transport des protéines
17.
Cell Chem Biol ; 23(2): 205-207, 2016 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-26971869

RÉSUMÉ

Necroptosis is a form of regulated cell death implicated in a range of inflammatory diseases. In this issue of Cell Chemical Biology, Li et al. (2016) describe a novel inhibitor of necroptosis that targets the chaperone protein HSP90.


Sujet(s)
Protéines du choc thermique HSP90/métabolisme , Nécrose/métabolisme , Apoptose , Mort cellulaire , Lignée cellulaire tumorale , Survie cellulaire , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique HSP72/métabolisme , Humains , Chaperons moléculaires/métabolisme , Protein kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Transduction du signal
18.
PLoS One ; 9(12): e115592, 2014.
Article de Anglais | MEDLINE | ID: mdl-25526394

RÉSUMÉ

The ability of commensal bacteria to influence gene expression in host cells under the influence of pathogenic bacteria has previously been demonstrated, however the extent of this interaction is important for understanding how bacteria can be used as probiotics. Real-time quantitative polymerase chain reaction is the most sensitive tool for evaluating relative changes to gene expression levels. However as a result of its sensitivity an appropriate method of normalisation should be used to account for any variation incurred in preparatory experimental procedures. These variations may result from differences in the amount of starting material, quality of extracted RNA, or in the efficiency of the reverse transcriptase or polymerase enzymes. Selection of an endogenous control gene is the preferred method of normalisation, and ideally a proper validation of the gene's appropriateness for the study in question should be performed. In this study we used quantitative polymerase chain reaction data and applied four different algorithms (geNorm, BestKeeper, NormFinder, and comparative ΔCq) to evaluate eleven different genes as to their suitability as endogenous controls for use in studies involving colonic (HT-29) and vaginal (VK2/E6E7) human mucosal epithelial cells treated with probiotic and pathogenic bacteria. We found phosphoglycerate kinase 1 to be most appropriate for HT-29 cells, and ribosomal protein large P0 to be the best choice for VK2/E6E7 cells. We also showed that use of less stable reference genes can lead to less accurate quantification of expression levels of gene of interest (GOI) and also can result in decreased statistical significance for GOI expression levels when compared to control. Additionally, we found the cell type being analysed had greater influence on reference gene selection than the treatment performed. This study provides recommendations for stable endogenous control genes for use in further studies involving colonic and vaginal cell lines after bacterial challenge.


Sujet(s)
Côlon/cytologie , Cellules épithéliales/microbiologie , Réaction de polymérisation en chaîne/normes , Vagin/cytologie , Lignée cellulaire , Cellules épithéliales/cytologie , Femelle , Analyse de profil d'expression de gènes/normes , Cellules HT29 , Humains , Réaction de polymérisation en chaîne/méthodes , Normes de référence
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...