165 results on '"Prescott AR"'
Search Results
2. The coated pit and macropinocytic pathways serve distinct endosome populations
- Author
-
Hewlett, LJ, primary, Prescott, AR, additional, and Watts, C, additional
- Published
- 1994
- Full Text
- View/download PDF
3. Artificial targeting of autophagy components to mitochondria reveals both conventional and unconventional mitophagy pathways.
- Author
-
Lorentzen KC, Prescott AR, and Ganley IG
- Abstract
Macroautophagy/autophagy enables lysosomal degradation of a diverse array of intracellular material. This process is essential for normal cellular function and its dysregulation is implicated in many diseases. Given this, there is much interest in understanding autophagic mechanisms of action in order to determine how it can be best targeted therapeutically. In mitophagy, the selective degradation of mitochondria via autophagy, mitochondria first need to be primed with signals that allow the recruitment of the core autophagy machinery to drive the local formation of an autophagosome around the target mitochondrion. To determine how the recruitment of different core autophagy components can drive mitophagy, we took advantage of the mito -QC mitophagy assay (an outer mitochondrial membrane-localized tandem mCherry-GFP tag). By tagging autophagy proteins with an anti-mCherry (or anti-GFP) nanobody, we could recruit them to mitochondria and simultaneously monitor levels of mitophagy. We found that targeting ULK1, ATG16L1 and the different Atg8-family proteins was sufficient to induce mitophagy. Mitochondrial recruitment of ULK1 and the Atg8-family proteins induced a conventional mitophagy pathway, requiring RB1CC1/FIP200, PIK3C3/VPS34 activity and ATG5. Surprisingly, the mitophagy pathway upon recruitment of ATG16L1 proceeded independently of ATG5, although it still required RB1CC1 and PIK3C3/VPS34 activity. In this latter pathway, mitochondria were alternatively delivered to lysosomes via uptake into early endosomes. Abbreviation: aGFP: anti-GFP nanobody; amCh: anti-mCherry nanobody; ATG: autophagy related; ATG16L1: autophagy related 16 like 1; AUTAC/AUTOTAC: autophagy-targeting chimera; BafA1: bafilomycin A
1 ; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CCCP: carbonyl cyanide m-chlorophenylhydrazone; COX4/COX IV: cytochrome c oxidase subunit 4; DFP: deferiprone; DMSO: dimethyl sulfoxide; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; HSPD1/HSP60: heat shock protein family D (Hsp60) member 1; HRP: horseradish peroxidase; HTRA2/OMI: HtrA serine peptidase 2; IB: immunoblotting; IF: immunofluorescence; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; NBR1: NBR1 autophagy cargo receptor; OMM: outer mitochondrial membrane; OPA1: OPA1 mitochondrial dynamin like GTPase; OPTN: optineurin; (D)PBS: (Dulbecco's) phosphate-buffered saline; PD: Parkinson disease; PFA: paraformaldehyde; POI: protein of interest; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; RAB: RAB, member RAS oncogene family; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SQSTM1: sequestosome 1; TAX1BP1: Tax1 binding protein 1; ULK: unc-51 like autophagy activating kinase 1; VPS: vacuolar protein sorting; WIPI: WD repeat domain, phosphoinositide interacting.- Published
- 2024
- Full Text
- View/download PDF
4. Additional feedforward mechanism of Parkin activation via binding of phospho-UBL and RING0 in trans .
- Author
-
Lenka DR, Dahe SV, Antico O, Sahoo P, Prescott AR, Muqit MMK, and Kumar A
- Subjects
- Humans, Phosphorylation, Crystallography, X-Ray, Models, Molecular, Ubiquitin metabolism, Kinetics, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases chemistry, Protein Binding
- Abstract
Loss-of-function Parkin mutations lead to early-onset of Parkinson's disease. Parkin is an auto-inhibited ubiquitin E3 ligase activated by dual phosphorylation of its ubiquitin-like (Ubl) domain and ubiquitin by the PINK1 kinase. Herein, we demonstrate a competitive binding of the phospho-Ubl and RING2 domains towards the RING0 domain, which regulates Parkin activity. We show that phosphorylated Parkin can complex with native Parkin, leading to the activation of autoinhibited native Parkin in trans . Furthermore, we show that the activator element (ACT) of Parkin is required to maintain the enzyme kinetics, and the removal of ACT slows the enzyme catalysis. We also demonstrate that ACT can activate Parkin in trans but less efficiently than when present in the cis molecule. Furthermore, the crystal structure reveals a donor ubiquitin binding pocket in the linker connecting REP and RING2, which plays a crucial role in Parkin activity., Competing Interests: DL, SD, OA, PS, AP, AK No competing interests declared, MM MM. is a member of the Scientific Advisory Board of Montara Therapeutics Inc and a scientific consultant to MSD UK, (© 2024, Lenka et al.)
- Published
- 2024
- Full Text
- View/download PDF
5. PINK1 regulated mitophagy is evident in skeletal muscles.
- Author
-
Singh F, Wilhelm L, Prescott AR, Ostacolo K, Zhao JF, Ogmundsdottir MH, and Ganley IG
- Abstract
PINK1, mutated in familial forms of Parkinson's disease, initiates mitophagy following mitochondrial depolarization. However, it is difficult to monitor this pathway physiologically in mice as loss of PINK1 does not alter basal mitophagy levels in most tissues. To further characterize this pathway in vivo , we used mito -QC mice in which loss of PINK1 was combined with the mitochondrial-associated POLG
D257A mutation. We focused on skeletal muscle as gene expression data indicates that this tissue has the highest PINK1 levels. We found that loss of PINK1 in oxidative hindlimb muscle significantly reduced mitophagy. Of interest, the presence of the POLGD257A mutation, while having a minor effect in most tissues, restored levels of muscle mitophagy caused by the loss of PINK1. Although our observations highlight that multiple mitophagy pathways operate within a single tissue, we identify skeletal muscle as a tissue of choice for the study of PINK1-dependant mitophagy under basal conditions., Competing Interests: Disclosure statement IGG is a consultant for Mitobridge Inc.- Published
- 2024
- Full Text
- View/download PDF
6. Golgi-IP, a tool for multimodal analysis of Golgi molecular content.
- Author
-
Fasimoye R, Dong W, Nirujogi RS, Rawat ES, Iguchi M, Nyame K, Phung TK, Bagnoli E, Prescott AR, Alessi DR, and Abu-Remaileh M
- Subjects
- Humans, Chromatography, Liquid, Lipids, Uridine Diphosphate metabolism, Golgi Apparatus metabolism, Proteome metabolism
- Abstract
The Golgi is a membrane-bound organelle that is essential for protein and lipid biosynthesis. It represents a central trafficking hub that sorts proteins and lipids to various destinations or for secretion from the cell. The Golgi has emerged as a docking platform for cellular signaling pathways including LRRK2 kinase whose deregulation leads to Parkinson disease. Golgi dysfunction is associated with a broad spectrum of diseases including cancer, neurodegeneration, and cardiovascular diseases. To allow the study of the Golgi at high resolution, we report a rapid Golgi immunoprecipitation technique (Golgi-IP) to isolate intact Golgi mini-stacks for subsequent analysis of their content. By fusing the Golgi-resident protein TMEM115 to three tandem HA epitopes (GolgiTAG), we purified the Golgi using Golgi-IP with minimal contamination from other compartments. We then established an analysis pipeline using liquid chromatography coupled with mass spectrometry to characterize the human Golgi proteome, metabolome, and lipidome. Subcellular proteomics confirmed known Golgi proteins and identified proteins not previously associated with the Golgi. Metabolite profiling established the human Golgi metabolome and revealed the enrichment of uridine-diphosphate (UDP) sugars and their derivatives, which is consistent with their roles in protein and lipid glycosylation. Furthermore, targeted metabolomics validated SLC35A2 as the subcellular transporter for UDP-hexose. Finally, lipidomics analysis showed that phospholipids including phosphatidylcholine, phosphatidylinositol, and phosphatidylserine are the most abundant Golgi lipids and that glycosphingolipids are enriched in this compartment. Altogether, our work establishes a comprehensive molecular map of the human Golgi and provides a powerful method to study the Golgi with high precision in health and disease.
- Published
- 2023
- Full Text
- View/download PDF
7. Correction: TPL2-mediated activation of ERK1 and ERK2 regulates the processing of pre-TNFα in LPS-stimulated macrophages.
- Author
-
Rousseau S, Papoutsopoulou M, Symons A, Cook D, Lucocq JM, Prescott AR, O'Garra A, Ley SC, and Cohen P
- Published
- 2022
- Full Text
- View/download PDF
8. Impact of 100 LRRK2 variants linked to Parkinson's disease on kinase activity and microtubule binding.
- Author
-
Kalogeropulou AF, Purlyte E, Tonelli F, Lange SM, Wightman M, Prescott AR, Padmanabhan S, Sammler E, and Alessi DR
- Subjects
- Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Microtubules metabolism, Mutation, Phosphorylation, Protein Binding, Parkinson Disease genetics, Parkinson Disease metabolism
- Abstract
Mutations enhancing the kinase activity of leucine-rich repeat kinase-2 (LRRK2) cause Parkinson's disease (PD) and therapies that reduce LRRK2 kinase activity are being tested in clinical trials. Numerous rare variants of unknown clinical significance have been reported, but how the vast majority impact on LRRK2 function is unknown. Here, we investigate 100 LRRK2 variants linked to PD, including previously described pathogenic mutations. We identify 23 LRRK2 variants that robustly stimulate kinase activity, including variants within the N-terminal non-catalytic regions (ARM (E334K, A419V), ANK (R767H), LRR (R1067Q, R1325Q)), as well as variants predicted to destabilize the ROC:CORB interface (ROC (A1442P, V1447M), CORA (R1628P) CORB (S1761R, L1795F)) and COR:COR dimer interface (CORB (R1728H/L)). Most activating variants decrease LRRK2 biomarker site phosphorylation (pSer935/pSer955/pSer973), consistent with the notion that the active kinase conformation blocks their phosphorylation. We conclude that the impact of variants on kinase activity is best evaluated by deploying a cellular assay of LRRK2-dependent Rab10 substrate phosphorylation, compared with a biochemical kinase assay, as only a minority of activating variants (CORB (Y1699C, R1728H/L, S1761R) and kinase (G2019S, I2020T, T2031S)), enhance in vitro kinase activity of immunoprecipitated LRRK2. Twelve variants including several that activate LRRK2 and have been linked to PD, suppress microtubule association in the presence of a Type I kinase inhibitor (ARM (M712V), LRR (R1320S), ROC (A1442P, K1468E, S1508R), CORA (A1589S), CORB (Y1699C, R1728H/L) and WD40 (R2143M, S2350I, G2385R)). Our findings will stimulate work to better understand the mechanisms by which variants impact biology and provide rationale for variant carrier inclusion or exclusion in ongoing and future LRRK2 inhibitor clinical trials., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
9. Differential roles and regulation of the protein kinases PAK4, PAK5 and PAK6 in melanoma cells.
- Author
-
Murugesan G, Prescott AR, Toth R, Campbell DG, Wells CM, and MacKintosh C
- Subjects
- Humans, Phorbol Esters, Phosphorylation, p21-Activated Kinases genetics, p21-Activated Kinases metabolism, Melanoma genetics, Protein Kinases metabolism
- Abstract
The protein kinases PAK4, PAK5 and PAK6 comprise a family of ohnologues. In multiple cancers including melanomas PAK5 most frequently carries non-synonymous mutations; PAK6 and PAK4 have fewer; and PAK4 is often amplified. To help interpret these genomic data, initially we compared the cellular regulation of the sister kinases and their roles in melanoma cells. In common with many ohnologue protein kinases, PAK4, PAK5 and PAK6 each have two 14-3-3-binding phosphosites of which phosphoSer99 is conserved. PAK4 localises to the leading edge of cells in response to phorbol ester-stimulated binding of 14-3-3 to phosphoSer99 and phosphoSer181, which are phosphorylated by two different PKCs or PKDs. These phosphorylations of PAK4 are essential for its phorbol ester-stimulated phosphorylation of downstream substrates. In contrast, 14-3-3 interacts with PAK5 in response to phorbol ester-stimulated phosphorylation of Ser99 and epidermal growth factor-stimulated phosphorylation of Ser288; whereas PAK6 docks onto 14-3-3 and is prevented from localising to cell-cell junctions when Ser133 is phosphorylated in response to cAMP-elevating agents via PKA and insulin-like growth factor 1 via PKB/Akt. Silencing of PAK4 impairs viability, migration and invasive behaviour of melanoma cells carrying BRAFV600E or NRASQ61K mutations. These defects are rescued by ectopic expression of PAK4, more so by a 14-3-3-binding deficient PAK4, and barely by PAK5 or PAK6. Together these genomic, biochemical and cellular data suggest that the oncogenic properties of PAK4 are regulated by PKC-PKD signalling in melanoma, while PAK5 and PAK6 are dispensable in this cancer., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. Condensation properties of stress granules and processing bodies are compromised in myotonic dystrophy type 1.
- Author
-
Gulyurtlu S, Magon MS, Guest P, Papavasiliou PP, Morrison KD, Prescott AR, and Sleeman JE
- Subjects
- Animals, Mammals metabolism, Myotonin-Protein Kinase genetics, Myotonin-Protein Kinase metabolism, Processing Bodies, RNA, Stress Granules, Trinucleotide Repeat Expansion genetics, Myotonic Dystrophy genetics
- Abstract
RNA regulation in mammalian cells requires complex physical compartmentalisation, using structures thought to be formed by liquid-liquid phase separation. Disruption of these structures is implicated in numerous degenerative diseases. Myotonic dystrophy type 1 (DM1) is a multi-systemic trinucleotide repeat disorder resulting from an expansion of nucleotides CTG (CTGexp) in the DNA encoding DM1 protein kinase (DMPK). The cellular hallmark of DM1 is the formation of nuclear foci that contain expanded DMPK RNA (CUGexp) (with thymine instead of uracil). We report here the deregulation of stress granules (SGs) and processing bodies (P-bodies), two cytoplasmic structures key for mRNA regulation, in cell culture models of DM1. Alterations to the rates of formation and dispersal of SGs suggest an altered ability of cells to respond to stress associated with DM1, while changes to the structure and dynamics of SGs and P-bodies suggest that a widespread alteration to the biophysical properties of cellular structures is a consequence of the presence of CUGexp RNA., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
11. Publisher's Note: ZNRF2 is released from membranes by growth factors and, together with ZNRF1, regulates the Na+/K+ATPase.
- Author
-
Hoxhaj G, Najafov A, Toth R, Campbell DG, Prescott AR, and MacKintosh C
- Published
- 2022
- Full Text
- View/download PDF
12. Why are the phenotypes of TRAF6 knock-in and TRAF6 knock-out mice so different?
- Author
-
Petrova T, Bennett K, Nanda S, Strickson S, Scudamore CL, Prescott AR, and Cohen P
- Subjects
- Animals, Autoimmune Diseases genetics, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes metabolism, Dermatitis genetics, Dermatitis pathology, Gene Knock-In Techniques, Mice, Mice, Knockout, Phenotype, Pneumonia genetics, Pneumonia pathology, Signal Transduction, Autoimmune Diseases pathology, Receptors, Interleukin-1 Type I metabolism, TNF Receptor-Associated Factor 6 genetics, TNF Receptor-Associated Factor 6 metabolism, Toll-Like Receptors metabolism
- Abstract
The expression of TNF-Receptor Associated Factor 6 (TRAF6) is essential for many physiological processes. Here we studied the phenotype of TRAF6[L74H] knock-in mice which are devoid of TRAF6 E3 ligase activity in every cell of the body, but express normal levels of the TRAF6 protein. Remarkably, TRAF6[L74H] mice have none of the phenotypes seen in TRAF6 KO mice. Instead TRAF6[L74H] mice display an entirely different phenotype, exhibiting autoimmunity, and severe inflammation of the skin and modest inflammation of the liver and lungs. Similar to mice with a Treg-specific knockout of TRAF6, or mice devoid of TRAF6 in all T cells, the CD4+ and CD8+ T cells in the spleen and lymph nodes displayed an activated effector memory phenotype with CD44high/CD62Llow expression on the cell surface. In contrast, T cells from WT mice exhibited the CD44low/CD62Lhigh phenotype characteristic of naïve T cells. The onset of autoimmunity and autoinflammation in TRAF6[L74H] mice (two weeks) was much faster than in mice with a Treg-specific knockout of TRAF6 or lacking TRAF6 expression in all T cells (2-3 months) and we discuss whether this may be caused by secondary inflammation of other tissues. The distinct phenotypes of mice lacking TRAF6 expression in all cells appears to be explained by their inability to signal via TNF Receptor Superfamily members, which does not seem to be impaired significantly in TRAF6[L74H] mice., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
13. The importance of the epithelial fibre cell interface to lens regeneration in an in vivo rat model and in a human bag-in-the-lens (BiL) sample.
- Author
-
Wu W, Lois N, Prescott AR, Brown AP, Van Gerwen V, Tassignon MJ, Richards SA, Saunter CD, Jarrin M, and Quinlan RA
- Subjects
- Actins metabolism, Aged, Animals, Aquaporins metabolism, Cadherins metabolism, Cell Proliferation physiology, Epithelial Cells ultrastructure, Epithelial-Mesenchymal Transition physiology, Eye Proteins metabolism, Female, Fibronectins metabolism, Humans, In Situ Nick-End Labeling, Lens Capsule, Crystalline cytology, Lens Capsule, Crystalline surgery, Lens, Crystalline ultrastructure, Male, Microscopy, Electron, Microscopy, Fluorescence, Models, Animal, Nerve Tissue Proteins metabolism, Proteomics, Rats, Rats, Sprague-Dawley, Tandem Mass Spectrometry, Capsule Opacification metabolism, Epithelial Cells physiology, Lens Implantation, Intraocular, Lens, Crystalline physiology, Regeneration physiology
- Abstract
Human lens regeneration and the Bag-in-the-Lens (BIL) surgical treatment for cataract both depend upon lens capsule closure for their success. Our studies suggest that the first three days after surgery are critical to their long-term outcomes. Using a rat model of lens regeneration, we evidenced lens epithelial cell (LEC) proliferation increased some 50 fold in the first day before rapidly declining to rates observed in the germinative zone of the contra-lateral, un-operated lens. Cell multi-layering at the lens equator occurred on days 1 and 2, but then reorganised into two discrete layers by day 3. E- and N-cadherin expression preceded cell polarity being re-established during the first week. Aquaporin 0 (AQP0) was first detected in the elongated cells at the lens equator at day 7. Cells at the capsulotomy site, however, behaved very differently expressing the epithelial mesenchymal transition (EMT) markers fibronectin and alpha-smooth muscle actin (SMA) from day 3 onwards. The physical interaction between the apical surfaces of the anterior and posterior LECs from day 3 after surgery preceded cell elongation. In the human BIL sample fibre cell formation was confirmed by both histological and proteome analyses, but the cellular response is less ordered and variable culminating in Soemmerring's ring (SR) formation and sometimes Elschnig's pearls. This we evidence for lenses from a single patient. No bow region or recognisable epithelial-fibre cell interface (EFI) was evident and consequently the fibre cells were disorganised. We conclude that lens cells require spatial and cellular cues to initiate, sustain and produce an optically functional tissue in addition to capsule integrity and the EFI., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
14. IKKβ is required for the formation of the NLRP3 inflammasome.
- Author
-
Nanda SK, Prescott AR, Figueras-Vadillo C, and Cohen P
- Subjects
- I-kappa B Kinase, Interleukin-1beta, Lipopolysaccharides, Nigericin, trans-Golgi Network, Inflammasomes genetics, NLR Family, Pyrin Domain-Containing 3 Protein genetics
- Abstract
The rapid formation and activation of the NLRP3 inflammasome is induced by co-stimulation with LPS and nigericin. It requires the LPS-stimulated activation of IKKβ, which exerts its effects independently of de novo gene transcription, protein translation and other protein kinases activated by IKKβ. IKKβ is not required for the nigericin-induced dispersion of the trans-Golgi network (TGN), but to bring NLRP3 in proximity with TGN38. The nigericin-induced dispersion of the Golgi is enhanced by co-stimulation with LPS, and this enhancement is IKKβ-dependent. Prolonged stimulation with LPS to increase the expression of NLRP3, followed by stimulation with nigericin, produced larger TGN38-positive puncta, and the ensuing activation of the NLRP3 inflammasome was also suppressed by IKKβ inhibitors added prior to stimulation with nigericin. IKKβ therefore has a key role in recruiting NLRP3 to the dispersed TGN, leading to the formation and activation of the NLRP3 inflammasome., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
15. Pharmacological rescue of impaired mitophagy in Parkinson's disease-related LRRK2 G2019S knock-in mice.
- Author
-
Singh F, Prescott AR, Rosewell P, Ball G, Reith AD, and Ganley IG
- Subjects
- Animals, Female, Male, Mice, Mice, Knockout, Mice, Transgenic, Mitophagy drug effects, Mutation, Parkinson Disease genetics, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 antagonists & inhibitors, Mitophagy genetics, Parkinson Disease physiopathology
- Abstract
Parkinson's disease (PD) is a major and progressive neurodegenerative disorder, yet the biological mechanisms involved in its aetiology are poorly understood. Evidence links this disorder with mitochondrial dysfunction and/or impaired lysosomal degradation - key features of the autophagy of mitochondria, known as mitophagy. Here, we investigated the role of LRRK2, a protein kinase frequently mutated in PD, in this process in vivo. Using mitophagy and autophagy reporter mice, bearing either knockout of LRRK2 or expressing the pathogenic kinase-activating G2019S LRRK2 mutation, we found that basal mitophagy was specifically altered in clinically relevant cells and tissues. Our data show that basal mitophagy inversely correlates with LRRK2 kinase activity in vivo. In support of this, use of distinct LRRK2 kinase inhibitors in cells increased basal mitophagy, and a CNS penetrant LRRK2 kinase inhibitor, GSK3357679A, rescued the mitophagy defects observed in LRRK2 G2019S mice. This study provides the first in vivo evidence that pathogenic LRRK2 directly impairs basal mitophagy, a process with strong links to idiopathic Parkinson's disease, and demonstrates that pharmacological inhibition of LRRK2 is a rational mitophagy-rescue approach and potential PD therapy., Competing Interests: FS, AP, PR, GB, AR, IG No competing interests declared, (© 2021, Singh et al.)
- Published
- 2021
- Full Text
- View/download PDF
16. Nucleotide sugar biosynthesis occurs in the glycosomes of procyclic and bloodstream form Trypanosoma brucei.
- Author
-
Sampaio Guther ML, Prescott AR, Kuettel S, Tinti M, and Ferguson MAJ
- Subjects
- Life Cycle Stages physiology, Microbodies enzymology, Trypanosoma brucei brucei enzymology, Microbodies metabolism, Nucleotides biosynthesis, Sugars metabolism, Trypanosoma brucei brucei metabolism
- Abstract
In Trypanosoma brucei, there are fourteen enzymatic biotransformations that collectively convert glucose into five essential nucleotide sugars: UDP-Glc, UDP-Gal, UDP-GlcNAc, GDP-Man and GDP-Fuc. These biotransformations are catalyzed by thirteen discrete enzymes, five of which possess putative peroxisome targeting sequences. Published experimental analyses using immunofluorescence microscopy and/or digitonin latency and/or subcellular fractionation and/or organelle proteomics have localized eight and six of these enzymes to the glycosomes of bloodstream form and procyclic form T. brucei, respectively. Here we increase these glycosome localizations to eleven in both lifecycle stages while noting that one, phospho-N-acetylglucosamine mutase, also localizes to the cytoplasm. In the course of these studies, the heterogeneity of glycosome contents was also noted. These data suggest that, unlike other eukaryotes, all of nucleotide sugar biosynthesis in T. brucei is compartmentalized to the glycosomes in both lifecycle stages. The implications are discussed., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
17. Inhibition of IL-34 Unveils Tissue-Selectivity and Is Sufficient to Reduce Microglial Proliferation in a Model of Chronic Neurodegeneration.
- Author
-
Obst J, Simon E, Martin-Estebane M, Pipi E, Barkwill LM, Gonzalez-Rivera I, Buchanan F, Prescott AR, Faust D, Fox S, Brownlees J, Taylor D, Perry VH, Nuthall H, Atkinson PJ, Karran E, Routledge C, and Gomez-Nicola D
- Subjects
- Animals, Antibodies, Monoclonal toxicity, Antibodies, Neutralizing toxicity, Brain metabolism, Brain pathology, Cell Line, Tumor, Disease Models, Animal, Genes, fms, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Interleukins metabolism, Mice, Inbred C57BL, Mice, Transgenic, Microglia metabolism, Microglia pathology, Prion Diseases metabolism, Prion Diseases pathology, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Signal Transduction, Antibodies, Monoclonal pharmacology, Antibodies, Neutralizing pharmacology, Brain drug effects, Cell Proliferation drug effects, Interleukins antagonists & inhibitors, Microglia drug effects, Nerve Degeneration, Prion Diseases drug therapy
- Abstract
The proliferation and activation of microglia, the resident macrophages in the brain, is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and prion disease. Colony stimulating factor 1 receptor (CSF1R) is critically involved in regulating microglial proliferation, and CSF1R blocking strategies have been recently used to modulate microglia in neurodegenerative diseases. However, CSF1R is broadly expressed by many cell types and the impact of its inhibition on the innate immune system is still unclear. CSF1R can be activated by two independent ligands, CSF-1 and interleukin 34 (IL-34). Recently, it has been reported that microglia development and maintenance depend on IL-34 signaling. In this study, we evaluate the inhibition of IL-34 as a novel strategy to reduce microglial proliferation in the ME7 model of prion disease. Selective inhibition of IL-34 showed no effects on peripheral macrophage populations in healthy mice, avoiding the side effects observed after CSF1R inhibition on the systemic compartment. However, we observed a reduction in microglial proliferation after IL-34 inhibition in prion-diseased mice, indicating that microglia could be more specifically targeted by reducing IL-34. Overall, our results highlight the challenges of targeting the CSF1R/IL34 axis in the systemic and central compartments, important for framing any therapeutic effort to tackle microglia/macrophage numbers during brain disease., (Copyright © 2020 Obst, Simon, Martin-Estebane, Pipi, Barkwill, Gonzalez-Rivera, Buchanan, Prescott, Faust, Fox, Brownlees, Taylor, Perry, Nuthall, Atkinson, Karran, Routledge and Gomez-Nicola.)
- Published
- 2020
- Full Text
- View/download PDF
18. Antibody RING-Mediated Destruction of Endogenous Proteins.
- Author
-
Ibrahim AFM, Shen L, Tatham MH, Dickerson D, Prescott AR, Abidi N, Xirodimas DP, and Hay RT
- Subjects
- Endopeptidases immunology, HeLa Cells, Humans, NEDD8 Protein immunology, Nuclear Proteins immunology, Proteasome Endopeptidase Complex immunology, Proteolysis, Single-Domain Antibodies immunology, Transcription Factors immunology, Ubiquitination, Endopeptidases metabolism, NEDD8 Protein metabolism, Nuclear Proteins metabolism, Proteasome Endopeptidase Complex metabolism, Single-Domain Antibodies metabolism, Transcription Factors metabolism, Ubiquitin metabolism
- Abstract
To understand gene function, the encoding DNA or mRNA transcript can be manipulated and the consequences observed. However, these approaches do not have a direct effect on the protein product of the gene, which is either permanently abrogated or depleted at a rate defined by the half-life of the protein. We therefore developed a single-component system that could induce the rapid degradation of the specific endogenous protein itself. A construct combining the RING domain of ubiquitin E3 ligase RNF4 with a protein-specific camelid nanobody mediates target destruction by the ubiquitin proteasome system, a process we describe as antibody RING-mediated destruction (ARMeD). The technique is highly specific because we observed no off-target protein destruction. Furthermore, bacterially produced nanobody-RING fusion proteins electroporated into cells induce degradation of target within minutes. With increasing availability of protein-specific nanobodies, this method will allow rapid and specific degradation of a wide range of endogenous proteins., Competing Interests: Declaration of Interests The authors declare no competing financial interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
19. Turtles and Tortoises Are in Trouble.
- Author
-
Stanford CB, Iverson JB, Rhodin AGJ, Paul van Dijk P, Mittermeier RA, Kuchling G, Berry KH, Bertolero A, Bjorndal KA, Blanck TEG, Buhlmann KA, Burke RL, Congdon JD, Diagne T, Edwards T, Eisemberg CC, Ennen JR, Forero-Medina G, Frankel M, Fritz U, Gallego-García N, Georges A, Gibbons JW, Gong S, Goode EV, Shi HT, Hoang H, Hofmeyr MD, Horne BD, Hudson R, Juvik JO, Kiester RA, Koval P, Le M, Lindeman PV, Lovich JE, Luiselli L, McCormack TEM, Meyer GA, Páez VP, Platt K, Platt SG, Pritchard PCH, Quinn HR, Roosenburg WM, Seminoff JA, Shaffer HB, Spencer R, Van Dyke JU, Vogt RC, and Walde AD
- Subjects
- Animals, Endangered Species, Extinction, Biological, Population Dynamics, Conservation of Natural Resources, Turtles
- Abstract
Turtles and tortoises (chelonians) have been integral components of global ecosystems for about 220 million years and have played important roles in human culture for at least 400,000 years. The chelonian shell is a remarkable evolutionary adaptation, facilitating success in terrestrial, freshwater and marine ecosystems. Today, more than half of the 360 living species and 482 total taxa (species and subspecies combined) are threatened with extinction. This places chelonians among the groups with the highest extinction risk of any sizeable vertebrate group. Turtle populations are declining rapidly due to habitat loss, consumption by humans for food and traditional medicines and collection for the international pet trade. Many taxa could become extinct in this century. Here, we examine survival threats to turtles and tortoises and discuss the interventions that will be needed to prevent widespread extinction in this group in coming decades., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
20. Cold climate adaptation is a plausible cause for evolution of multicellular sporulation in Dictyostelia.
- Author
-
Lawal HM, Schilde C, Kin K, Brown MW, James J, Prescott AR, and Schaap P
- Subjects
- Acclimatization, Biological Evolution, Cold Climate, Phylogeny, Spores physiology, Dictyostelium classification, Dictyostelium physiology, Fossils parasitology
- Abstract
Unicellular protozoa that encyst individually upon starvation evolved at least eight times into organisms that instead form multicellular fruiting bodies with spores. The Dictyostelia are the largest and most complex group of such organisms. They can be subdivided into 4 major groups, with many species in groups 1-3 having additionally retained encystment. To understand fitness differences between spores and cysts, we measured long-term survival of spores and cysts under climate-mimicking conditions, investigated spore and cyst ultrastructure, and related fitness characteristics to species ecology. We found that spores and cysts survived 22 °C equally well, but that spores survived wet and dry frost better than cysts, with group 4 spores being most resilient. Spore walls consist of three layers and those of cysts of maximally two, while spores were also more compacted than cysts, with group 4 spores being the most compacted. Group 4 species were frequently isolated from arctic and alpine zones, which was rarely the case for group 1-3 species. We inferred a fossil-calibrated phylogeny of Dictyostelia, which showed that its two major branches diverged 0.52 billion years ago, following several global glaciations. Our results suggest that Dictyostelium multicellular sporulation was a likely adaptation to a cold climate.
- Published
- 2020
- Full Text
- View/download PDF
21. A conserved ATG2-GABARAP family interaction is critical for phagophore formation.
- Author
-
Bozic M, van den Bekerom L, Milne BA, Goodman N, Roberston L, Prescott AR, Macartney TJ, Dawe N, and McEwan DG
- Subjects
- Apoptosis Regulatory Proteins metabolism, Autophagy, Autophagy-Related Proteins genetics, Autophagy-Related Proteins metabolism, Humans, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism, Protein Transport, Vesicular Transport Proteins metabolism, Autophagosomes metabolism, Membrane Proteins metabolism
- Abstract
The intracellular trafficking pathway, macroautophagy, is a recycling and disposal service that can be upregulated during periods of stress to maintain cellular homeostasis. An essential phase is the elongation and closure of the phagophore to seal and isolate unwanted cargo prior to lysosomal degradation. Human ATG2A and ATG2B proteins, through their interaction with WIPI proteins, are thought to be key players during phagophore elongation and closure, but little mechanistic detail is known about their function. We have identified a highly conserved motif driving the interaction between human ATG2 and GABARAP proteins that is in close proximity to the ATG2-WIPI4 interaction site. We show that the ATG2A-GABARAP interaction mutants are unable to form and close phagophores resulting in blocked autophagy, similar to ATG2A/ATG2B double-knockout cells. In contrast, the ATG2A-WIPI4 interaction mutant fully restored phagophore formation and autophagy flux, similar to wild-type ATG2A. Taken together, we provide new mechanistic insights into the requirements for ATG2 function at the phagophore and suggest that an ATG2-GABARAP/GABARAP-L1 interaction is essential for phagophore formation, whereas ATG2-WIPI4 interaction is dispensable., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
22. Functional and proteomic analysis of a full thickness filaggrin-deficient skin organoid model.
- Author
-
Elias MS, Wright SC, Nicholson WV, Morrison KD, Prescott AR, Ten Have S, Whitfield PD, Lamond AI, and Brown SJ
- Abstract
Background: Atopic eczema is an itchy inflammatory disorder characterised by skin barrier dysfunction. Loss-of-function mutations in the gene encoding filaggrin ( FLG ) are a major risk factor, but the mechanisms by which filaggrin haploinsufficiency leads to atopic inflammation remain incompletely understood. Skin as an organ that can be modelled using primary cells in vitro provides the opportunity for selected genetic effects to be investigated in detail. Methods: Primary human keratinocytes and donor-matched primary fibroblasts from healthy individuals were used to create skin organoid models with and without siRNA-mediated knockdown of FLG . Biological replicate sets of organoids were assessed using histological, functional and biochemical measurements. Results: FLG knockdown leads to subtle changes in histology and ultrastructure including a reduction in thickness of the stratum corneum and smaller, less numerous keratohyalin granules. Immature organoids showed some limited evidence of barrier impairment with FLG knockdown, but the mature organoids showed no difference in transepidermal water loss, water content or dye penetration. There was no difference in epidermal ceramide content. Mass spectrometry proteomic analysis detected >8000 proteins per sample. Gene ontology and pathway analyses identified an increase in transcriptional and translational activity but a reduction in proteins contributing to terminal differentiation, including caspase 14, dermokine, AKT1 and TGF-beta-1. Aspects of innate and adaptive immunity were represented in both the up-regulated and down-regulated protein groups, as was the term 'axon guidance'. Conclusions: This work provides further evidence for keratinocyte-specific mechanisms contributing to immune and neurological, as well as structural, aspects of skin barrier dysfunction. Individuals with filaggrin deficiency may derive benefit from future therapies targeting keratinocyte-immune crosstalk and neurogenic pruritus., Competing Interests: No competing interests were disclosed., (Copyright: © 2019 Elias MS et al.)
- Published
- 2019
- Full Text
- View/download PDF
23. Phosphoproteomics reveals that the hVPS34 regulated SGK3 kinase specifically phosphorylates endosomal proteins including Syntaxin-7, Syntaxin-12, RFIP4 and WDR44.
- Author
-
Malik N, Nirujogi RS, Peltier J, Macartney T, Wightman M, Prescott AR, Gourlay R, Trost M, Alessi DR, and Karapetsas A
- Subjects
- Biomarkers metabolism, Cell Membrane metabolism, Gene Knockout Techniques, HEK293 Cells, Heterocyclic Compounds, 3-Ring pharmacology, Humans, Insulin-Like Growth Factor I pharmacology, Phosphorylation drug effects, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Qa-SNARE Proteins genetics, Substrate Specificity, Transfection, Class III Phosphatidylinositol 3-Kinases metabolism, Endosomes metabolism, Protein Serine-Threonine Kinases metabolism, Qa-SNARE Proteins metabolism
- Abstract
The serum- and glucocorticoid-regulated kinase (SGK) isoforms contribute resistance to cancer therapies targeting the PI3K pathway. SGKs are homologous to Akt and these kinases display overlapping specificity and phosphorylate several substrates at the same residues, such as TSC2 to promote tumor growth by switching on the mTORC1 pathway. The SGK3 isoform is up-regulated in breast cancer cells treated with PI3K or Akt inhibitors and recruited and activated at endosomes, through its phox homology domain binding to PtdIns(3)P. We undertook genetic and pharmacological phosphoproteomic screens to uncover novel SGK3 substrates. We identified 40 potential novel SGK3 substrates, including four endosomal proteins STX7 (Ser126) and STX12 (Ser139), RFIP4 (Ser527) and WDR44 (Ser346) that were efficiently phosphorylated in vitro by SGK3 at the sites identified in vivo, but poorly by Akt. We demonstrate that these substrates are inefficiently phosphorylated by Akt as they possess an n + 1 residue from the phosphorylation site that is unfavorable for Akt phosphorylation. Phos-tag analysis revealed that stimulation of HEK293 cells with IGF1 to activate SGK3, promoted phosphorylation of a significant fraction of endogenous STX7 and STX12, in a manner that was blocked by knock-out of SGK3 or treatment with a pan SGK inhibitor (14H). SGK3 phosphorylation of STX12 enhanced interaction with the VAMP4/VTI1A/STX6 containing the SNARE complex and promoted plasma membrane localization. Our data reveal novel substrates for SGK3 and suggest a mechanism by which STX7 and STX12 SNARE complexes are regulated by SGK3. They reveal new biomarkers for monitoring SGK3 pathway activity., (© 2019 The Author(s).)
- Published
- 2019
- Full Text
- View/download PDF
24. Proteomic analysis of a filaggrin-deficient skin organoid model shows evidence of increased transcriptional-translational activity, keratinocyte-immune crosstalk and disordered axon guidance.
- Author
-
Elias MS, Wright SC, Nicholson WV, Morrison KD, Prescott AR, Ten Have S, Whitfield PD, Lamond AI, and Brown SJ
- Abstract
Background: Atopic eczema is an itchy inflammatory disorder characterised by skin barrier dysfunction. Loss-of-function mutations in the gene encoding filaggrin ( FLG ) are a major risk factor, but the mechanisms by which filaggrin haploinsufficiency leads to atopic inflammation remain incompletely understood. Skin as an organ that can be modelled using primary cells in vitro provides the opportunity for selected genetic effects to be investigated in detail. Methods: Primary human keratinocytes and donor-matched primary fibroblasts from healthy individuals were used to create skin organoid models with and without siRNA-mediated knockdown of FLG . Biological replicate sets of organoids were assessed using histological, functional and biochemical measurements. Results: FLG knockdown leads to subtle changes in histology and ultrastructure including a reduction in thickness of the stratum corneum and smaller, less numerous keratohyalin granules. Immature organoids showed evidence of barrier impairment with FLG knockdown, but the mature organoids showed no difference in transepidermal water loss, water content or dye penetration. There was no difference in epidermal ceramide content. Mass spectrometry proteomic analysis detected >8000 proteins per sample. Gene ontology and pathway analyses identified an increase in transcriptional and translational activity but a reduction in proteins contributing to terminal differentiation, including caspase 14, dermokine, AKT1 and TGF-beta-1. Aspects of innate and adaptive immunity were represented in both the up-regulated and down-regulated protein groups, as was the term 'axon guidance'. Conclusions: This work provides further evidence for keratinocyte-specific mechanisms contributing to immune and neurological, as well as structural, aspects of skin barrier dysfunction. Individuals with filaggrin deficiency may derive benefit from future therapies targeting keratinocyte-immune crosstalk and neurogenic pruritus., Competing Interests: No competing interests were disclosed., (Copyright: © 2019 Elias MS et al.)
- Published
- 2019
- Full Text
- View/download PDF
25. FAM83D directs protein kinase CK1α to the mitotic spindle for proper spindle positioning.
- Author
-
Fulcher LJ, He Z, Mei L, Macartney TJ, Wood NT, Prescott AR, Whigham AJ, Varghese J, Gourlay R, Ball G, Clarke R, Campbell DG, Maxwell CA, and Sapkota GP
- Subjects
- Animals, Casein Kinase I genetics, Cell Cycle genetics, Cell Cycle physiology, Cell Cycle Proteins genetics, Cell Line, Tumor, Flow Cytometry, HeLa Cells, Humans, Mice, Mice, Knockout, Microtubule-Associated Proteins genetics, Mitosis genetics, Mitosis physiology, Casein Kinase I metabolism, Cell Cycle Proteins metabolism, Microtubule-Associated Proteins metabolism, Spindle Apparatus metabolism
- Abstract
The concerted action of many protein kinases helps orchestrate the error-free progression through mitosis of mammalian cells. The roles and regulation of some prominent mitotic kinases, such as cyclin-dependent kinases, are well established. However, these and other known mitotic kinases alone cannot account for the extent of protein phosphorylation that has been reported during mammalian mitosis. Here we demonstrate that CK1α, of the casein kinase 1 family of protein kinases, localises to the spindle and is required for proper spindle positioning and timely cell division. CK1α is recruited to the spindle by FAM83D, and cells devoid of FAM83D, or those harbouring CK1α-binding-deficient FAM83D
F283A/F283A knockin mutations, display pronounced spindle positioning defects, and a prolonged mitosis. Restoring FAM83D at the endogenous locus in FAM83D-/- cells, or artificially delivering CK1α to the spindle in FAM83DF283A/F283A cells, rescues these defects. These findings implicate CK1α as new mitotic kinase that orchestrates the kinetics and orientation of cell division., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2019
- Full Text
- View/download PDF
26. EMSY expression affects multiple components of the skin barrier with relevance to atopic dermatitis.
- Author
-
Elias MS, Wright SC, Remenyi J, Abbott JC, Bray SE, Cole C, Edwards S, Gierlinski M, Glok M, McGrath JA, Nicholson WV, Paternoster L, Prescott AR, Have ST, Whitfield PD, Lamond AI, and Brown SJ
- Subjects
- Chromosomes, Human, Pair 11 genetics, Chromosomes, Human, Pair 11 immunology, Dermatitis, Atopic genetics, Dermatitis, Atopic pathology, Female, Filaggrin Proteins, Genome-Wide Association Study, Humans, Male, Membrane Proteins genetics, Membrane Proteins immunology, Neoplasm Proteins genetics, Nuclear Proteins genetics, Repressor Proteins genetics, Skin pathology, Dermatitis, Atopic immunology, Gene Expression Regulation immunology, Neoplasm Proteins immunology, Nuclear Proteins immunology, Repressor Proteins immunology, Skin immunology, Transcription, Genetic immunology
- Abstract
Background: Atopic dermatitis (AD) is a common, complex, and highly heritable inflammatory skin disease. Genome-wide association studies offer opportunities to identify molecular targets for drug development. A risk locus on chromosome 11q13.5 lies between 2 candidate genes, EMSY and LRRC32 (leucine-rich repeat-containing 32) but the functional mechanisms affecting risk of AD remain unclear., Objectives: We sought to apply a combination of genomic and molecular analytic techniques to investigate which genes are responsible for genetic risk at this locus and to define mechanisms contributing to atopic skin disease., Methods: We used interrogation of available genomic and chromosome conformation data in keratinocytes, small interfering RNA (siRNA)-mediated knockdown in skin organotypic culture and functional assessment of barrier parameters, mass spectrometric global proteomic analysis and quantitative lipid analysis, electron microscopy of organotypic skin, and immunohistochemistry of human skin samples., Results: Genomic data indicate active promoters in the genome-wide association study locus and upstream of EMSY; EMSY, LRRC32, and intergenic variants all appear to be within a single topologically associating domain. siRNA-knockdown of EMSY in organotypic culture leads to enhanced development of barrier function, reflecting increased expression of structural and functional proteins, including filaggrin and filaggrin-2, as well as long-chain ceramides. Conversely, overexpression of EMSY in keratinocytes leads to a reduction in markers of barrier formation. Skin biopsy samples from patients with AD show greater EMSY staining in the nucleus, which is consistent with an increased functional effect of this transcriptional control protein., Conclusion: Our findings demonstrate an important role for EMSY in transcriptional regulation and skin barrier formation, supporting EMSY inhibition as a therapeutic approach., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
27. A comparative map of macroautophagy and mitophagy in the vertebrate eye.
- Author
-
McWilliams TG, Prescott AR, Villarejo-Zori B, Ball G, Boya P, and Ganley IG
- Subjects
- Animals, Autophagosomes metabolism, Cell Differentiation physiology, Ciliary Body cytology, Ciliary Body metabolism, Cornea cytology, Cornea metabolism, Eye cytology, Eye growth & development, Homeostasis physiology, Lens, Crystalline cytology, Lens, Crystalline metabolism, Lysosomes metabolism, Mice, Mice, Inbred C57BL, Mitochondria metabolism, Optic Nerve cytology, Optic Nerve metabolism, Photoreceptor Cells, Vertebrate cytology, Photoreceptor Cells, Vertebrate metabolism, Retina cytology, Retina metabolism, Eye metabolism, Macroautophagy physiology, Mitophagy physiology
- Abstract
Photoreception is pivotal to our experience and perception of the natural world; hence the eye is of prime importance for most vertebrate animals to sense light. Central to visual health is mitochondrial homeostasis, and the selective autophagic turnover of mitochondria (mitophagy) is predicted to play a key role here. Despite studies that link aberrant mitophagy to ocular dysfunction, little is known about the prevalence of basal mitophagy, or its relationship to general autophagy, in the visual system. In this study, we utilize the mito -QC mouse and a closely related general macroautophagy reporter model to profile basal mitophagy and macroautophagy in the adult and developing eye. We report that ocular macroautophagy is widespread, but surprisingly mitophagy does not always follow the same pattern of occurrence. We observe low levels of mitophagy in the lens and ciliary body, in stark contrast to the high levels of general MAP1LC3-dependent macroautophagy in these regions. We uncover a striking reversal of this process in the adult retina, where mitophagy accounts for a larger degree of the macroautophagy taking place, specifically in the photoreceptor neurons of the outer nuclear layer. We also show the developmental regulation of autophagy in a variety of ocular tissues. In particular, mitophagy in the adult mouse retina is reversed in localization during the latter stages of development. Our work thus defines the landscape of mitochondrial homeostasis in the mammalian eye, and in doing so highlights the selective nature of autophagy in vivo and the specificity of the reporters used. Abbreviations: ATG: autophagy related; GFP: green fluorescent protein; LC3: microtubule associated protein 1 light chain 3; ONH: optic nerve head; ONL: outer nuclear layer; RPE: retinal pigment epithelium.
- Published
- 2019
- Full Text
- View/download PDF
28. Rapid and Reversible Knockdown of Endogenously Tagged Endosomal Proteins via an Optimized HaloPROTAC Degrader.
- Author
-
Tovell H, Testa A, Maniaci C, Zhou H, Prescott AR, Macartney T, Ciulli A, and Alessi DR
- Subjects
- CRISPR-Cas Systems, Class III Phosphatidylinositol 3-Kinases genetics, Gene Knock-In Techniques, HEK293 Cells, Humans, Protein Serine-Threonine Kinases genetics, Endosomes metabolism, Gene Editing methods, Proteins metabolism
- Abstract
Inducing post-translational protein knockdown is an important approach to probe biology and validate drug targets. An efficient strategy to achieve this involves expression of a protein of interest fused to an exogenous tag, allowing tag-directed chemical degraders to mediate protein ubiquitylation and proteasomal degradation. Here, we combine improved HaloPROTAC degrader probes with CRISPR/Cas9 genome editing technology to trigger rapid degradation of endogenous target proteins. Our optimized probe, HaloPROTAC-E, a chloroalkane conjugate of high-affinity VHL binder VH298, induced reversible degradation of two endosomally localized proteins, SGK3 and VPS34, with a DC
50 of 3-10 nM. HaloPROTAC-E induced rapid (∼50% degradation after 30 min) and complete ( Dmax of ∼95% at 48 h) depletion of Halo-tagged SGK3, blocking downstream phosphorylation of the SGK3 substrate NDRG1. HaloPROTAC-E more potently induced greater steady state degradation of Halo tagged endogenous VPS34 than the previously reported HaloPROTAC3 compound. Quantitative global proteomics revealed that HaloPROTAC-E is remarkably selective inducing only degradation of the Halo tagged endogenous VPS34 complex (VPS34, VPS15, Beclin1, and ATG14) and no other proteins were significantly degraded. This study exemplifies the combination of HaloPROTACs with CRISPR/Cas9 endogenous protein tagging as a useful method to induce rapid and reversible degradation of endogenous proteins to interrogate their function.- Published
- 2019
- Full Text
- View/download PDF
29. Phosphorylation of Parkin at serine 65 is essential for its activation in vivo .
- Author
-
McWilliams TG, Barini E, Pohjolan-Pirhonen R, Brooks SP, Singh F, Burel S, Balk K, Kumar A, Montava-Garriga L, Prescott AR, Hassoun SM, Mouton-Liger F, Ball G, Hills R, Knebel A, Ulusoy A, Di Monte DA, Tamjar J, Antico O, Fears K, Smith L, Brambilla R, Palin E, Valori M, Eerola-Rautio J, Tienari P, Corti O, Dunnett SB, Ganley IG, Suomalainen A, and Muqit MMK
- Subjects
- Animals, Humans, Mice, Mice, Transgenic, Mitochondria genetics, Mitochondria pathology, Parkinson Disease genetics, Parkinson Disease pathology, Phosphorylation genetics, Protein Kinases genetics, Serine genetics, Serine metabolism, Mitochondria metabolism, Mitophagy, Parkinson Disease metabolism, Protein Kinases metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Mutations in PINK1 and Parkin result in autosomal recessive Parkinson's disease (PD). Cell culture and in vitro studies have elaborated the PINK1-dependent regulation of Parkin and defined how this dyad orchestrates the elimination of damaged mitochondria via mitophagy. PINK1 phosphorylates ubiquitin at serine 65 (Ser65) and Parkin at an equivalent Ser65 residue located within its N-terminal ubiquitin-like domain, resulting in activation; however, the physiological significance of Parkin Ser65 phosphorylation in vivo in mammals remains unknown. To address this, we generated a Parkin Ser65Ala (S65A) knock-in mouse model. We observe endogenous Parkin Ser65 phosphorylation and activation in mature primary neurons following mitochondrial depolarization and reveal this is disrupted in Parkin
S65A/S65A neurons. Phenotypically, ParkinS65A/S65A mice exhibit selective motor dysfunction in the absence of any overt neurodegeneration or alterations in nigrostriatal mitophagy. The clinical relevance of our findings is substantiated by the discovery of homozygous PARKIN ( PARK2 ) p.S65N mutations in two unrelated patients with PD. Moreover, biochemical and structural analysis demonstrates that the ParkinS65N/S65N mutant is pathogenic and cannot be activated by PINK1. Our findings highlight the central role of Parkin Ser65 phosphorylation in health and disease., (© 2018 The Authors.)- Published
- 2018
- Full Text
- View/download PDF
30. Basal Mitophagy Occurs Independently of PINK1 in Mouse Tissues of High Metabolic Demand.
- Author
-
McWilliams TG, Prescott AR, Montava-Garriga L, Ball G, Singh F, Barini E, Muqit MMK, Brooks SP, and Ganley IG
- Subjects
- Animals, Dopaminergic Neurons metabolism, Mice, Knockout, Microglia metabolism, Stress, Physiological, Mitophagy, Organ Specificity, Protein Kinases metabolism
- Abstract
Dysregulated mitophagy has been linked to Parkinson's disease (PD) due to the role of PTEN-induced kinase 1 (PINK1) in mediating depolarization-induced mitophagy in vitro. Elegant mouse reporters have revealed the pervasive nature of basal mitophagy in vivo, yet the role of PINK1 and tissue metabolic context remains unknown. Using mito-QC, we investigated the contribution of PINK1 to mitophagy in metabolically active tissues. We observed a high degree of mitophagy in neural cells, including PD-relevant mesencephalic dopaminergic neurons and microglia. In all tissues apart from pancreatic islets, loss of Pink1 did not influence basal mitophagy, despite disrupting depolarization-induced Parkin activation. Our findings provide the first in vivo evidence that PINK1 is detectable at basal levels and that basal mammalian mitophagy occurs independently of PINK1. This suggests multiple, yet-to-be-discovered pathways orchestrating mammalian mitochondrial integrity in a context-dependent fashion, and this has profound implications for our molecular understanding of vertebrate mitophagy., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Mechanism of activation of SGK3 by growth factors via the Class 1 and Class 3 PI3Ks.
- Author
-
Malik N, Macartney T, Hornberger A, Anderson KE, Tovell H, Prescott AR, and Alessi DR
- Subjects
- Class I Phosphatidylinositol 3-Kinases metabolism, Class III Phosphatidylinositol 3-Kinases metabolism, Endosomes drug effects, Endosomes metabolism, Gene Expression Regulation, HEK293 Cells, Humans, Insulin-Like Growth Factor I metabolism, Mechanistic Target of Rapamycin Complex 2 metabolism, Phosphatidylinositol Phosphates metabolism, Phosphorylation drug effects, Plasmids chemistry, Plasmids metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Transfection, Class I Phosphatidylinositol 3-Kinases genetics, Class III Phosphatidylinositol 3-Kinases genetics, Insulin-Like Growth Factor I pharmacology, Mechanistic Target of Rapamycin Complex 2 genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Derailment of the PI3K-AGC protein kinase signalling network contributes to many human diseases including cancer. Recent work has revealed that the poorly studied AGC kinase family member, SGK3, promotes resistance to cancer therapies that target the Class 1 PI3K pathway, by substituting for loss of Akt kinase activity. SGK3 is recruited and activated at endosomes, by virtue of its phox homology domain binding to PtdIns(3)P. Here, we demonstrate that endogenous SGK3 is rapidly activated by growth factors such as IGF1, through pathways involving both Class 1 and Class 3 PI3Ks. We provide evidence that IGF1 enhances endosomal PtdIns(3)P levels via a pathway involving the UV-RAG complex of hVPS34 Class 3 PI3K. Our data point towards IGF1-induced activation of Class 1 PI3K stimulating SGK3 through enhanced production of PtdIns(3)P resulting from the dephosphorylation of PtdIns(3,4,5)P
3 Our findings are also consistent with activation of Class 1 PI3K promoting mTORC2 phosphorylation of SGK3 and with oncogenic Ras-activating SGK3 solely through the Class 1 PI3K pathway. Our results highlight the versatility of upstream pathways that activate SGK3 and help explain how SGK3 substitutes for Akt following inhibition of Class 1 PI3K/Akt pathways. They also illustrate robustness of SGK3 activity that can remain active and counteract physiological conditions or stresses where either Class 1 or Class 3 PI3K pathways are inhibited., (© 2018 The Author(s).)- Published
- 2018
- Full Text
- View/download PDF
32. Bifunctionality of a biofilm matrix protein controlled by redox state.
- Author
-
Arnaouteli S, Ferreira AS, Schor M, Morris RJ, Bromley KM, Jo J, Cortez KL, Sukhodub T, Prescott AR, Dietrich LEP, MacPhee CE, and Stanley-Wall NR
- Subjects
- Bacillus subtilis genetics, Bacterial Proteins chemistry, Bacterial Proteins metabolism, Hydrophobic and Hydrophilic Interactions, Oxidation-Reduction, Bacillus subtilis physiology, Bacterial Proteins physiology, Biofilms
- Abstract
Biofilms are communities of microbial cells that are encapsulated within a self-produced polymeric matrix. The matrix is critical to the success of biofilms in diverse habitats; however, many details of the composition, structure, and function remain enigmatic. Biofilms formed by the Gram-positive bacterium Bacillus subtilis depend on the production of the secreted film-forming protein BslA. Here, we show that a gradient of electron acceptor availability through the depth of the biofilm gives rise to two distinct functional roles for BslA and that these roles can be genetically separated through targeted amino acid substitutions. We establish that monomeric BslA is necessary and sufficient to give rise to complex biofilm architecture, whereas dimerization of BslA is required to render the community hydrophobic. Dimerization of BslA, mediated by disulfide bond formation, depends on two conserved cysteine residues located in the C-terminal region. Our findings demonstrate that bacteria have evolved multiple uses for limited elements in the matrix, allowing for alternative responses in a complex, changing environment., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
33. Brd4-Brd2 isoform switching coordinates pluripotent exit and Smad2-dependent lineage specification.
- Author
-
Fernandez-Alonso R, Davidson L, Hukelmann J, Zengerle M, Prescott AR, Lamond A, Ciulli A, Sapkota GP, and Findlay GM
- Subjects
- Animals, Cell Cycle Proteins, Cell Line, Cell Lineage, Humans, Mice, Proteins metabolism, Signal Transduction, Cell Differentiation, Nuclear Proteins metabolism, Pluripotent Stem Cells metabolism, Protein Serine-Threonine Kinases metabolism, Smad2 Protein metabolism, Transcription Factors metabolism
- Abstract
Pluripotent stem cells (PSCs) hold great clinical potential, as they possess the capacity to differentiate into fully specialised tissues such as pancreas, liver, neurons and cardiac muscle. However, the molecular mechanisms that coordinate pluripotent exit with lineage specification remain poorly understood. To address this question, we perform a small molecule screen to systematically identify novel regulators of the Smad2 signalling network, a key determinant of PSC fate. We reveal an essential function for BET family bromodomain proteins in Smad2 activation, distinct from the role of Brd4 in pluripotency maintenance. Mechanistically, BET proteins specifically engage Nodal gene regulatory elements (NREs) to promote Nodal signalling and Smad2 developmental responses. In pluripotent cells, Brd2-Brd4 occupy NREs, but only Brd4 is required for pluripotency gene expression. Brd4 downregulation facilitates pluripotent exit and drives enhanced Brd2 NRE occupancy, thereby unveiling a specific function for Brd2 in differentiative Nodal-Smad2 signalling. Therefore, distinct BET functionalities and Brd4-Brd2 isoform switching at NREs coordinate pluripotent exit with lineage specification., (© 2017 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2017
- Full Text
- View/download PDF
34. mito-QC illuminates mitophagy and mitochondrial architecture in vivo.
- Author
-
McWilliams TG, Prescott AR, Allen GF, Tamjar J, Munson MJ, Thomson C, Muqit MM, and Ganley IG
- Subjects
- Animals, Cerebellum cytology, Embryo, Mammalian cytology, Female, Fibroblasts cytology, Fibroblasts metabolism, Genes, Reporter, Kidney Cortex cytology, Kidney Cortex metabolism, Kidney Tubules cytology, Kidney Tubules metabolism, Mammals metabolism, Mice, Transgenic, Neurons cytology, Neurons metabolism, Organ Specificity, Mitochondria metabolism, Mitophagy
- Abstract
Autophagic turnover of mitochondria, termed mitophagy, is proposed to be an essential quality-control (QC) mechanism of pathophysiological relevance in mammals. However, if and how mitophagy proceeds within specific cellular subtypes in vivo remains unclear, largely because of a lack of tractable tools and models. To address this, we have developed "mito-QC," a transgenic mouse with a pH-sensitive fluorescent mitochondrial signal. This allows the assessment of mitophagy and mitochondrial architecture in vivo. Using confocal microscopy, we demonstrate that mito-QC is compatible with classical and contemporary techniques in histochemistry and allows unambiguous in vivo detection of mitophagy and mitochondrial morphology at single-cell resolution within multiple organ systems. Strikingly, our model uncovers highly enriched and differential zones of mitophagy in the developing heart and within specific cells of the adult kidney. mito-QC is an experimentally advantageous tool of broad relevance to cell biology researchers within both discovery-based and translational research communities., (© 2016 McWilliams et al.)
- Published
- 2016
- Full Text
- View/download PDF
35. The E3 ubiquitin ligase ZNRF2 is a substrate of mTORC1 and regulates its activation by amino acids.
- Author
-
Hoxhaj G, Caddye E, Najafov A, Houde VP, Johnson C, Dissanayake K, Toth R, Campbell DG, Prescott AR, and MacKintosh C
- Subjects
- GTP Phosphohydrolases metabolism, Gene Knockdown Techniques, Humans, Mechanistic Target of Rapamycin Complex 1, Protein Interaction Mapping, Ubiquitin-Protein Ligases genetics, Vacuolar Proton-Translocating ATPases metabolism, Amino Acids metabolism, Gene Expression Regulation, Enzymologic, Multiprotein Complexes metabolism, TOR Serine-Threonine Kinases metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
The mechanistic Target of Rapamycin complex 1 (mTORC1) senses intracellular amino acid levels through an intricate machinery, which includes the Rag GTPases, Ragulator and vacuolar ATPase (V-ATPase). The membrane-associated E3 ubiquitin ligase ZNRF2 is released into the cytosol upon its phosphorylation by Akt. In this study, we show that ZNRF2 interacts with mTOR on membranes, promoting the amino acid-stimulated translocation of mTORC1 to lysosomes and its activation in human cells. ZNRF2 also interacts with the V-ATPase and preserves lysosomal acidity. Moreover, knockdown of ZNRF2 decreases cell size and cell proliferation. Upon growth factor and amino acid stimulation, mTORC1 phosphorylates ZNRF2 on Ser145, and this phosphosite is dephosphorylated by protein phosphatase 6. Ser145 phosphorylation stimulates vesicle-to-cytosol translocation of ZNRF2 and forms a novel negative feedback on mTORC1. Our findings uncover ZNRF2 as a component of the amino acid sensing machinery that acts upstream of Rag-GTPases and the V-ATPase to activate mTORC1.
- Published
- 2016
- Full Text
- View/download PDF
36. A key role for PTP1B in dendritic cell maturation, migration, and T cell activation.
- Author
-
Martin-Granados C, Prescott AR, Le Sommer S, Klaska IP, Yu T, Muckersie E, Giuraniuc CV, Grant L, Delibegovic M, and Forrester JV
- Subjects
- Animals, Bone Marrow Cells, Cell Differentiation, Cell Movement physiology, Cells, Cultured, Chemokine CCL19 metabolism, Coculture Techniques, Female, Mice, Mice, Knockout, Myeloid Cells enzymology, Nuclear Receptor Coactivator 1 metabolism, Protein Tyrosine Phosphatase, Non-Receptor Type 1 genetics, Receptors, CCR7 metabolism, STAT3 Transcription Factor metabolism, Dendritic Cells immunology, Lymphocyte Activation, Podosomes genetics, Protein Tyrosine Phosphatase, Non-Receptor Type 1 physiology, T-Lymphocytes immunology
- Abstract
Dendritic cells (DC) are the major antigen-presenting cells bridging innate and adaptive immunity, a function they perform by converting quiescent DC to active, mature DC with the capacity to activate naïve T cells. They do this by migrating from the tissues to the T cell area of the secondary lymphoid tissues. Here, we demonstrate that myeloid cell-specific genetic deletion of PTP1B (LysM PTP1B) leads to defects in lipopolysaccharide-driven bone marrow-derived DC (BMDC) activation associated with increased levels of phosphorylated Stat3. We show that myeloid cell-specific PTP1B deletion also causes decreased migratory capacity of epidermal DC, as well as reduced CCR7 expression and chemotaxis to CCL19 by BMDC. PTP1B deficiency in BMDC also impairs their migration in vivo. Further, immature LysM PTP1B BMDC display fewer podosomes, increased levels of phosphorylated Src at tyrosine 527, and loss of Src localization to podosome puncta. In co-culture with T cells, LysM PTP1B BMDC establish fewer and shorter contacts than control BMDC. Finally, LysM PTP1B BMDC fail to present antigen to T cells as efficiently as control BMDC. These data provide first evidence for a key regulatory role for PTP1B in mediating a central DC function of initiating adaptive immune responses in response to innate immune cell activation., (© The Author (2015). Published by Oxford University Press on behalf of Journal of Molecular Cell Biology, IBCB, SIBS, CAS. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
37. The PDK1-Rsk Signaling Pathway Controls Langerhans Cell Proliferation and Patterning.
- Author
-
Zaru R, Matthews SP, Edgar AJ, Prescott AR, Gomez-Nicola D, Hanauer A, and Watts C
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases genetics, Animals, Animals, Newborn, Cell Count, Cell Movement, Cell Size, Cells, Cultured, Epidermal Cells, Epidermis metabolism, Flow Cytometry, Langerhans Cells metabolism, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Confocal, Phosphorylation, Ribosomal Protein S6 Kinases, 90-kDa genetics, 3-Phosphoinositide-Dependent Protein Kinases metabolism, Cell Proliferation, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Signal Transduction
- Abstract
Langerhans cells (LC), the dendritic cells of the epidermis, are distributed in a distinctive regularly spaced array. In the mouse, the LC array is established in the first few days of life from proliferating local precursors, but the regulating signaling pathways are not fully understood. We found that mice lacking the kinase phosphoinositide-dependent kinase 1 selectively lack LC. Deletion of the phosphoinositide-dependent kinase 1 target kinases, ribosomal S6 kinase 1 (Rsk1) and Rsk2, produced a striking perturbation in the LC network: LC density was reduced 2-fold, but LC size was increased by the same magnitude. Reduced LC numbers in Rsk1/2(-/-) mice was not due to accelerated emigration from the skin but rather to reduced proliferation at least in adults. Rsk1/2 were required for normal LC patterning in neonates, but not when LC were ablated in adults and replaced by bone marrow-derived cells. Increased LC size was an intrinsic response to reduced LC numbers, reversible on LC emigration, and could be observed in wild type epidermis where LC size also correlated inversely with LC density. Our results identify a key signaling pathway needed to establish a normal LC network and suggest that LC might maintain epidermal surveillance by increasing their "footprint" when their numbers are limited., (Copyright © 2015 The Authors.)
- Published
- 2015
- Full Text
- View/download PDF
38. High-resolution quantitative proteome analysis reveals substantial differences between phagosomes of RAW 264.7 and bone marrow derived macrophages.
- Author
-
Guo M, Härtlova A, Dill BD, Prescott AR, Gierliński M, and Trost M
- Subjects
- Animals, Cells, Cultured, Mice, Protein Interaction Maps, Proteomics, Databases, Protein, Macrophages chemistry, Phagosomes chemistry, Proteome analysis, Proteome chemistry, Proteome classification, RAW 264.7 Cells chemistry, RAW 264.7 Cells cytology
- Abstract
Macrophages are important immune cells operating at the forefront of innate immunity by taking up foreign particles and microbes through phagocytosis. The RAW 264.7 cell line is commonly used for experiments in the macrophage and phagocytosis field. However, little is known how its functions compare to primary macrophages. Here, we have performed an in-depth proteomics characterization of phagosomes from RAW 264.7 and bone marrow derived macrophages by quantifying more than 2500 phagosomal proteins. Our data indicate that there are significant differences for a large number of proteins including important receptors such as mannose receptor 1 and Siglec-1. Moreover, bone marrow derived macrophages phagosomes mature considerably faster by fusion with endosomes and the lysosome which we validated using fluorogenic phagocytic assays. We provide a valuable resource for researcher in the field and recommend careful use of the RAW 264.7 cell line when studying phagosome functions. All MS data have been deposited in the ProteomeXchange with identifier PXD001293 (http://proteomecentral.proteomexchange.org/dataset/PXD001293)., (© 2014 The Authors. PROTEOMICS published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
39. Binding to serine 65-phosphorylated ubiquitin primes Parkin for optimal PINK1-dependent phosphorylation and activation.
- Author
-
Kazlauskaite A, Martínez-Torres RJ, Wilkie S, Kumar A, Peltier J, Gonzalez A, Johnson C, Zhang J, Hope AG, Peggie M, Trost M, van Aalten DM, Alessi DR, Prescott AR, Knebel A, Walden H, and Muqit MM
- Subjects
- HEK293 Cells, Humans, Mass Spectrometry, Mutagenesis, Site-Directed, Mutation, Phosphorylation, Protein Binding, Protein Kinases genetics, Protein Structure, Tertiary, Serine genetics, Ubiquitin genetics, Ubiquitin-Protein Ligases genetics, Ubiquitination, Protein Kinases metabolism, Serine metabolism, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Mutations in the mitochondrial protein kinase PINK1 are associated with autosomal recessive Parkinson disease (PD). We and other groups have reported that PINK1 activates Parkin E3 ligase activity both directly via phosphorylation of Parkin serine 65 (Ser(65))--which lies within its ubiquitin-like domain (Ubl)--and indirectly through phosphorylation of ubiquitin at Ser(65). How Ser(65)-phosphorylated ubiquitin (ubiquitin(Phospho-Ser65)) contributes to Parkin activation is currently unknown. Here, we demonstrate that ubiquitin(Phospho-Ser65) binding to Parkin dramatically increases the rate and stoichiometry of Parkin phosphorylation at Ser(65) by PINK1 in vitro. Analysis of the Parkin structure, corroborated by site-directed mutagenesis, shows that the conserved His302 and Lys151 residues play a critical role in binding of ubiquitin(Phospho-Ser65), thereby promoting Parkin Ser(65) phosphorylation and activation of its E3 ligase activity in vitro. Mutation of His302 markedly inhibits Parkin Ser(65) phosphorylation at the mitochondria, which is associated with a marked reduction in its E3 ligase activity following mitochondrial depolarisation. We show that the binding of ubiquitin(Phospho-Ser65) to Parkin disrupts the interaction between the Ubl domain and C-terminal region, thereby increasing the accessibility of Parkin Ser(65). Finally, purified Parkin maximally phosphorylated at Ser(65) in vitro cannot be further activated by the addition of ubiquitin(Phospho-Ser65). Our results thus suggest that a major role of ubiquitin(Phospho-Ser65) is to promote PINK1-mediated phosphorylation of Parkin at Ser(65), leading to maximal activation of Parkin E3 ligase activity. His302 and Lys151 are likely to line a phospho-Ser(65)-binding pocket on the surface of Parkin that is critical for the ubiquitin(Phospho-Ser65) interaction. This study provides new mechanistic insights into Parkin activation by ubiquitin(Phospho-Ser65), which could aid in the development of Parkin activators that mimic the effect of ubiquitin(Phospho-Ser65)., (© 2015 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
40. The clinically approved drugs dasatinib and bosutinib induce anti-inflammatory macrophages by inhibiting the salt-inducible kinases.
- Author
-
Ozanne J, Prescott AR, and Clark K
- Subjects
- Animals, Arginase immunology, Cells, Cultured, Cyclic AMP Response Element-Binding Protein immunology, Cytokines immunology, Dasatinib, Macrophages cytology, Mice, Phosphotransferases (Alcohol Group Acceptor) immunology, Protein Serine-Threonine Kinases immunology, Transcription Factors immunology, Tumor Necrosis Factor Ligand Superfamily Member 14 immunology, Aniline Compounds pharmacology, Immunity, Innate drug effects, Macrophages immunology, Nitriles pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrimidines pharmacology, Quinolines pharmacology, Thiazoles pharmacology
- Abstract
Macrophages switch to an anti-inflammatory, 'regulatory'-like phenotype characterized by the production of high levels of interleukin (IL)-10 and low levels of pro-inflammatory cytokines to promote the resolution of inflammation. A potential therapeutic strategy for the treatment of chronic inflammatory diseases would be to administer drugs that could induce the formation of 'regulatory'-like macrophages at sites of inflammation. In the present study, we demonstrate that the clinically approved cancer drugs bosutinib and dasatinib induce several hallmark features of 'regulatory'-like macrophages. Treatment of macrophages with bosutinib or dasatinib elevates the production of IL-10 while suppressing the production of IL-6, IL-12p40 and tumour necrosis factor α (TNFα) in response to Toll-like receptor (TLR) stimulation. Moreover, macrophages treated with bosutinib or dasatinib express higher levels of markers of 'regulatory'-like macrophages including LIGHT, SPHK1 and arginase 1. Bosutinib and dasatinib were originally developed as inhibitors of the protein tyrosine kinases Bcr-Abl and Src but we show that, surprisingly, the effects of bosutinib and dasatinib on macrophage polarization are the result of the inhibition of the salt-inducible kinases. Consistent with the present finding, bosutinib and dasatinib induce the dephosphorylation of CREB-regulated transcription co-activator 3 (CRTC3) and its nuclear translocation where it induces a cAMP-response-element-binding protein (CREB)-dependent gene transcription programme including that of IL-10. Importantly, these effects of bosutinib and dasatinib on IL-10 gene expression are lost in macrophages expressing a drug-resistant mutant of salt-inducible kinase 2 (SIK2). In conclusion, our study identifies the salt-inducible kinases as major targets of bosutinib and dasatinib that mediate the effects of these drugs on the innate immune system and provides novel mechanistic insights into the anti-inflammatory properties of these drugs.
- Published
- 2015
- Full Text
- View/download PDF
41. Characterization of VPS34-IN1, a selective inhibitor of Vps34, reveals that the phosphatidylinositol 3-phosphate-binding SGK3 protein kinase is a downstream target of class III phosphoinositide 3-kinase.
- Author
-
Bago R, Malik N, Munson MJ, Prescott AR, Davies P, Sommer E, Shpiro N, Ward R, Cross D, Ganley IG, and Alessi DR
- Subjects
- Amino Acid Motifs, Cell Line, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Class I Phosphatidylinositol 3-Kinases metabolism, Class II Phosphatidylinositol 3-Kinases metabolism, Class III Phosphatidylinositol 3-Kinases metabolism, Endosomes metabolism, Humans, Hydrophobic and Hydrophilic Interactions, Inositol Polyphosphate 5-Phosphatases, Phosphatidylinositol Phosphates metabolism, Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases, Phosphoinositide-3 Kinase Inhibitors, Phosphoric Monoester Hydrolases metabolism, Phosphorylation, Protein Binding, Protein Structure, Tertiary, Aminopyridines pharmacology, Class III Phosphatidylinositol 3-Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases metabolism, Pyrimidines pharmacology
- Abstract
The Vps34 (vacuolar protein sorting 34) class III PI3K (phosphoinositide 3-kinase) phosphorylates PtdIns (phosphatidylinositol) at endosomal membranes to generate PtdIns(3)P that regulates membrane trafficking processes via its ability to recruit a subset of proteins possessing PtdIns(3)P-binding PX (phox homology) and FYVE domains. In the present study, we describe a highly selective and potent inhibitor of Vps34, termed VPS34-IN1, that inhibits Vps34 with 25 nM IC50 in vitro, but does not significantly inhibit the activity of 340 protein kinases or 25 lipid kinases tested that include all isoforms of class I as well as class II PI3Ks. Administration of VPS34-IN1 to cells induces a rapid dose-dependent dispersal of a specific PtdIns(3)P-binding probe from endosome membranes, within 1 min, without affecting the ability of class I PI3K to regulate Akt. Moreover, we explored whether SGK3 (serum- and glucocorticoid-regulated kinase-3), the only protein kinase known to interact specifically with PtdIns(3)P via its N-terminal PX domain, might be controlled by Vps34. Mutations disrupting PtdIns(3)P binding ablated SGK3 kinase activity by suppressing phosphorylation of the T-loop [PDK1 (phosphoinositide-dependent kinase 1) site] and hydrophobic motif (mammalian target of rapamycin site) residues. VPS34-IN1 induced a rapid ~50-60% loss of SGK3 phosphorylation within 1 min. VPS34-IN1 did not inhibit activity of the SGK2 isoform that does not possess a PtdIns(3)P-binding PX domain. Furthermore, class I PI3K inhibitors (GDC-0941 and BKM120) that do not inhibit Vps34 suppressed SGK3 activity by ~40%. Combining VPS34-IN1 and GDC-0941 reduced SGK3 activity ~80-90%. These data suggest SGK3 phosphorylation and hence activity is controlled by two pools of PtdIns(3)P. The first is produced through phosphorylation of PtdIns by Vps34 at the endosome. The second is due to the conversion of class I PI3K product, PtdIns(3,4,5)P3 into PtdIns(3)P, via the sequential actions of the PtdIns 5-phosphatases [SHIP1/2 (Src homology 2-domain-containing inositol phosphatase 1/2)] and PtdIns 4-phosphatase [INPP4B (inositol polyphosphate 4-phosphatase type II)]. VPS34-IN1 will be a useful probe to delineate physiological roles of the Vps34. Monitoring SGK3 phosphorylation and activity could be employed as a biomarker of Vps34 activity, in an analogous manner by which Akt is used to probe cellular class I PI3K activity. Combining class I (GDC-0941) and class III (VPS34-IN1) PI3K inhibitors could be used as a strategy to better analyse the roles and regulation of the elusive class II PI3K.
- Published
- 2014
- Full Text
- View/download PDF
42. Interplay between Polo kinase, LKB1-activated NUAK1 kinase, PP1βMYPT1 phosphatase complex and the SCFβTrCP E3 ubiquitin ligase.
- Author
-
Banerjee S, Zagórska A, Deak M, Campbell DG, Prescott AR, and Alessi DR
- Subjects
- AMP-Activated Protein Kinase Kinases, Amino Acid Sequence, Animals, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation, Gene Expression Regulation, HEK293 Cells, Humans, Molecular Sequence Data, Myosin-Light-Chain Phosphatase metabolism, Protein Binding, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, Repressor Proteins antagonists & inhibitors, Repressor Proteins metabolism, SKP Cullin F-Box Protein Ligases metabolism, Sequence Alignment, Signal Transduction, Ubiquitin-Protein Ligases metabolism, Polo-Like Kinase 1, Cell Cycle genetics, Cell Cycle Proteins genetics, Myosin-Light-Chain Phosphatase genetics, Protein Kinases genetics, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins genetics, Repressor Proteins genetics, SKP Cullin F-Box Protein Ligases genetics, Ubiquitin-Protein Ligases genetics
- Abstract
NUAK1 (NUAK family SnF1-like kinase-1) and NUAK2 protein kinases are activated by the LKB1 tumour suppressor and have been implicated in regulating multiple processes such as cell survival, senescence, adhesion and polarity. In the present paper we present evidence that expression of NUAK1 is controlled by CDK (cyclin-dependent kinase), PLK (Polo kinase) and the SCFβTrCP (Skp, Cullin and F-boxβTrCP) E3 ubiquitin ligase complex. Our data indicate that CDK phosphorylates NUAK1 at Ser445, triggering binding to PLK, which subsequently phosphorylates NUAK1 at two conserved non-catalytic serine residues (Ser476 and Ser480). This induces binding of NUAK1 to βTrCP, the substrate-recognition subunit of the SCFβTrCP E3 ligase, resulting in NUAK1 becoming ubiquitylated and degraded. We also show that NUAK1 and PLK1 are reciprocally controlled in the cell cycle. In G2-M-phase, when PLK1 is most active, NUAK1 levels are low and vice versa in S-phase, when PLK1 expression is low, NUAK1 is more highly expressed. Moreover, NUAK1 inhibitors (WZ4003 or HTH-01-015) suppress proliferation by reducing the population of cells in S-phase and mitosis, an effect that can be rescued by overexpression of a NUAK1 mutant in which Ser476 and Ser480 are mutated to alanine. Finally, previous work has suggested that NUAK1 phosphorylates and inhibits PP1βMYPT1 (where PP1 is protein phosphatase 1) and that a major role for the PP1βMYPT1 complex is to inhibit PLK1 by dephosphorylating its T-loop (Thr210). We demonstrate that activation of NUAK1 leads to a striking increase in phosphorylation of PLK1 at Thr210, an effect that is suppressed by NUAK1 inhibitors. Our data link NUAK1 to important cell-cycle signalling components (CDK, PLK and SCFβTrCP) and suggest that NUAK1 plays a role in stimulating S-phase, as well as PLK1 activity via its ability to regulate the PP1βMYPT1 phosphatase.
- Published
- 2014
- Full Text
- View/download PDF
43. Biochemical analysis of TssK, a core component of the bacterial Type VI secretion system, reveals distinct oligomeric states of TssK and identifies a TssK-TssFG subcomplex.
- Author
-
English G, Byron O, Cianfanelli FR, Prescott AR, and Coulthurst SJ
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins metabolism, Base Sequence, Membrane Proteins genetics, Membrane Proteins metabolism, Molecular Sequence Data, Protein Multimerization, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Serratia marcescens chemistry, Serratia marcescens genetics, Bacterial Proteins chemistry, Bacterial Secretion Systems genetics, Gene Expression Regulation, Bacterial, Membrane Proteins chemistry, Serratia marcescens metabolism
- Abstract
Gram-negative bacteria use the Type VI secretion system (T6SS) to inject toxic proteins into rival bacteria or eukaryotic cells. However, the mechanism of the T6SS is incompletely understood. In the present study, we investigated a conserved component of the T6SS, TssK, using the antibacterial T6SS of Serratia marcescens as a model system. TssK was confirmed to be essential for effector secretion by the T6SS. The native protein, although not an integral membrane protein, appeared to localize to the inner membrane, consistent with its presence within a membrane-anchored assembly. Recombinant TssK purified from S. marcescens was found to exist in several stable oligomeric forms, namely trimer, hexamer and higher-order species. Native-level purification of TssK identified TssF and TssG as interacting proteins. TssF and TssG, conserved T6SS components of unknown function, were required for T6SS activity, but not for correct localization of TssK. A complex containing TssK, TssF and TssG was subsequently purified in vitro, confirming that these three proteins form a new subcomplex within the T6SS. Our findings provide new insight into the T6SS assembly, allowing us to propose a model whereby TssK recruits TssFG into the membrane-associated T6SS complex and different oligomeric states of TssK may contribute to the dynamic mechanism of the system.
- Published
- 2014
- Full Text
- View/download PDF
44. Transcriptionally correlated subcellular dynamics of MBNL1 during lens development and their implication for the molecular pathology of myotonic dystrophy type 1.
- Author
-
Coleman SM, Prescott AR, and Sleeman JE
- Subjects
- Alternative Splicing genetics, Animals, Cells, Cultured, Epithelial Cells physiology, Humans, Molecular Dynamics Simulation, Subcellular Fractions physiology, Swine, Lens, Crystalline cytology, Lens, Crystalline growth & development, Myotonic Dystrophy genetics, Myotonic Dystrophy pathology, RNA-Binding Proteins genetics, Transcription, Genetic genetics
- Abstract
DM1 (myotonic dystrophy type 1) is caused by elongation of a CTG repeat in the DMPK (dystrophia myotonica-protein kinase) gene. mRNA transcripts containing these CUGexp (CUG expansion) repeats form accumulations, or foci, in the nucleus of the cell. The pathogenesis of DM1 is proposed to result from inappropriate patterns of alternative splicing caused by sequestration of the developmentally regulated alternative splicing factor MBNL1 (muscleblind-like 1) by these foci. Since eye lens cataract is a common feature of DM1 we have examined the distribution and dynamics of MBNL1 in lens epithelial cell lines derived from patients with DM1. The results of the present study demonstrate that only a small proportion of nuclear MBNL1 accumulates in CUGexp pre-mRNA foci. MBNL1 is, however, highly mobile and changes localization in response to altered transcription and splicing activity. Moreover, immunolocalization studies in lens sections suggest that a change in MBNL1 distribution is important during lens growth and differentiation. Although these data suggest that the loss of MBNL1 function due to accumulation in foci is an unlikely explanation for DM1 symptoms in the lens, they do demonstrate a strong relationship between the subcellular MBNL1 localization and pathways of cellular differentiation, providing an insight into the sensitivity of the lens to changes in MBNL1 distribution.
- Published
- 2014
- Full Text
- View/download PDF
45. Time-resolved quantitative proteomics implicates the core snRNP protein SmB together with SMN in neural trafficking.
- Author
-
Prescott AR, Bales A, James J, Trinkle-Mulcahy L, and Sleeman JE
- Subjects
- Dyneins metabolism, HeLa Cells, Humans, Microtubules metabolism, Neurons metabolism, Protein Transport, Proteomics, RNA Splicing, Time-Lapse Imaging, Transport Vesicles metabolism, Vesicular Transport Proteins metabolism, Neurites metabolism, Proteome metabolism, Survival of Motor Neuron 1 Protein metabolism, snRNP Core Proteins metabolism
- Abstract
The biogenesis of splicing snRNPs (small nuclear ribonucleoproteins) is a complex process, beginning and ending in the nucleus of the cell but including key stages that take place in the cytoplasm. In particular, the SMN (survival motor neuron) protein complex is required for addition of the core Sm proteins to the snRNP. Insufficiency of SMN results in the inherited neurodegenerative condition, spinal muscular atrophy (SMA). Details of the physical organization of the cytoplasmic stages of snRNP biogenesis are unknown. Here, we use time-resolved quantitative proteomics to identify proteins that associate preferentially with either newly assembled or mature splicing snRNPs. We identified highly mobile SmB protein-trafficking vesicles in neural cells, which are dependent on the cellular levels of SMN and SmB for their morphology and mobility. We propose that these represent a family of related vesicles, some of which play a role in snRNP biogenesis and some that might play more diverse roles in cellular RNA metabolism.
- Published
- 2014
- Full Text
- View/download PDF
46. Characterization of WZ4003 and HTH-01-015 as selective inhibitors of the LKB1-tumour-suppressor-activated NUAK kinases.
- Author
-
Banerjee S, Buhrlage SJ, Huang HT, Deng X, Zhou W, Wang J, Traynor R, Prescott AR, Alessi DR, and Gray NS
- Subjects
- AMP-Activated Protein Kinase Kinases, Animals, Cell Movement drug effects, Cell Proliferation drug effects, Cells, Cultured, Drug Evaluation, Preclinical, Enzyme Activation drug effects, HEK293 Cells, Humans, Mice, Neoplasms pathology, Protein Kinases genetics, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases physiology, Repressor Proteins agonists, Repressor Proteins genetics, Substrate Specificity, Tumor Suppressor Proteins physiology, Anilides pharmacology, Benzodiazepinones pharmacology, Heterocyclic Compounds, 4 or More Rings pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrimidines pharmacology, Repressor Proteins antagonists & inhibitors
- Abstract
The related NUAK1 and NUAK2 are members of the AMPK (AMP-activated protein kinase) family of protein kinases that are activated by the LKB1 (liver kinase B1) tumour suppressor kinase. Recent work suggests they play important roles in regulating key biological processes including Myc-driven tumorigenesis, senescence, cell adhesion and neuronal polarity. In the present paper we describe the first highly specific protein kinase inhibitors of NUAK kinases namely WZ4003 and HTH-01-015. WZ4003 inhibits both NUAK isoforms (IC50 for NUAK1 is 20 nM and for NUAK2 is 100 nM), whereas HTH-01-015 inhibits only NUAK1 (IC50 is 100 nM). These compounds display extreme selectivity and do not significantly inhibit the activity of 139 other kinases that were tested including ten AMPK family members. In all cell lines tested, WZ4003 and HTH-01-015 inhibit the phosphorylation of the only well-characterized substrate, MYPT1 (myosin phosphate-targeting subunit 1) that is phosphorylated by NUAK1 at Ser(445). We also identify a mutation (A195T) that does not affect basal NUAK1 activity, but renders it ~50-fold resistant to both WZ4003 and HTH-01-015. Consistent with NUAK1 mediating the phosphorylation of MYPT1 we find that in cells overexpressing drug-resistant NUAK1[A195T], but not wild-type NUAK1, phosphorylation of MYPT1 at Ser(445) is no longer suppressed by WZ4003 or HTH-01-015. We also demonstrate that administration of WZ4003 and HTH-01-015 to MEFs (mouse embryonic fibroblasts) significantly inhibits migration in a wound-healing assay to a similar extent as NUAK1-knockout. WZ4003 and HTH-01-015 also inhibit proliferation of MEFs to the same extent as NUAK1 knockout and U2OS cells to the same extent as NUAK1 shRNA knockdown. We find that WZ4003 and HTH-01-015 impaired the invasive potential of U2OS cells in a 3D cell invasion assay to the same extent as NUAK1 knockdown. The results of the present study indicate that WZ4003 and HTH-01-015 will serve as useful chemical probes to delineate the biological roles of the NUAK kinases.
- Published
- 2014
- Full Text
- View/download PDF
47. A synthetic system for expression of components of a bacterial microcompartment.
- Author
-
Sargent F, Davidson FA, Kelly CL, Binny R, Christodoulides N, Gibson D, Johansson E, Kozyrska K, Lado LL, MacCallum J, Montague R, Ortmann B, Owen R, Coulthurst SJ, Dupuy L, Prescott AR, and Palmer T
- Subjects
- Escherichia coli genetics, Genetic Vectors, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Salmonella genetics, Metabolic Networks and Pathways, Propylene Glycols metabolism, Salmonella enzymology, Salmonella metabolism
- Abstract
In general, prokaryotes are considered to be single-celled organisms that lack internal membrane-bound organelles. However, many bacteria produce proteinaceous microcompartments that serve a similar purpose, i.e. to concentrate specific enzymic reactions together or to shield the wider cytoplasm from toxic metabolic intermediates. In this paper, a synthetic operon encoding the key structural components of a microcompartment was designed based on the genes for the Salmonella propanediol utilization (Pdu) microcompartment. The genes chosen included pduA, -B, -J, -K, -N, -T and -U, and each was shown to produce protein in an Escherichia coli chassis. In parallel, a set of compatible vectors designed to express non-native cargo proteins was also designed and tested. Engineered hexa-His tags allowed isolation of the components of the microcompartments together with co-expressed, untagged, cargo proteins. Finally, an in vivo protease accessibility assay suggested that a PduD-GFP fusion could be protected from proteolysis when co-expressed with the synthetic microcompartment operon. This work gives encouragement that it may be possible to harness the genes encoding a non-native microcompartment for future biotechnological applications.
- Published
- 2013
- Full Text
- View/download PDF
48. Tmem79/Matt is the matted mouse gene and is a predisposing gene for atopic dermatitis in human subjects.
- Author
-
Saunders SP, Goh CS, Brown SJ, Palmer CN, Porter RM, Cole C, Campbell LE, Gierlinski M, Barton GJ, Schneider G, Balmain A, Prescott AR, Weidinger S, Baurecht H, Kabesch M, Gieger C, Lee YA, Tavendale R, Mukhopadhyay S, Turner SW, Madhok VB, Sullivan FM, Relton C, Burn J, Meggitt S, Smith CH, Allen MA, Barker JN, Reynolds NJ, Cordell HJ, Irvine AD, McLean WH, Sandilands A, and Fallon PG
- Subjects
- Animals, Dermatitis, Atopic immunology, Dermatitis, Atopic pathology, Filaggrin Proteins, Gene Expression, Humans, Male, Mice, Mutation, Phenotype, Physical Chromosome Mapping, Polymorphism, Single Nucleotide, Skin metabolism, Skin pathology, Dermatitis, Atopic genetics, Genetic Predisposition to Disease, Membrane Proteins genetics
- Abstract
Background: Atopic dermatitis (AD) is a major inflammatory condition of the skin caused by inherited skin barrier deficiency, with mutations in the filaggrin gene predisposing to development of AD. Support for barrier deficiency initiating AD came from flaky tail mice, which have a frameshift mutation in Flg and also carry an unknown gene, matted, causing a matted hair phenotype., Objective: We sought to identify the matted mutant gene in mice and further define whether mutations in the human gene were associated with AD., Methods: A mouse genetics approach was used to separate the matted and Flg mutations to produce congenic single-mutant strains for genetic and immunologic analysis. Next-generation sequencing was used to identify the matted gene. Five independently recruited AD case collections were analyzed to define associations between single nucleotide polymorphisms (SNPs) in the human gene and AD., Results: The matted phenotype in flaky tail mice is due to a mutation in the Tmem79/Matt gene, with no expression of the encoded protein mattrin in the skin of mutant mice. Matt(ft) mice spontaneously have dermatitis and atopy caused by a defective skin barrier, with mutant mice having systemic sensitization after cutaneous challenge with house dust mite allergens. Meta-analysis of 4,245 AD cases and 10,558 population-matched control subjects showed that a missense SNP, rs6684514, [corrected] in the human MATT gene has a small but significant association with AD., Conclusion: In mice mutations in Matt cause a defective skin barrier and spontaneous dermatitis and atopy. A common SNP in MATT has an association with AD in human subjects., (Copyright © 2013 American Academy of Allergy, Asthma & Immunology. Published by Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. p53 and cell cycle independent dysregulation of autophagy in chronic lymphocytic leukaemia.
- Author
-
Groves MJ, Johnson CE, James J, Prescott AR, Cunningham J, Haydock S, Pepper C, Fegan C, Pirrie L, Westwood NJ, Coates PJ, Ganley IG, and Tauro S
- Subjects
- Aged, Aged, 80 and over, Apoptosis drug effects, Apoptosis genetics, Autophagy drug effects, Autophagy genetics, Benzamides pharmacology, Caspase 3 genetics, Cell Cycle drug effects, Cell Cycle genetics, Cell Cycle physiology, Cell Proliferation drug effects, Humans, Interleukin-2 pharmacology, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Microtubule-Associated Proteins genetics, Middle Aged, Proteasome Endopeptidase Complex drug effects, Proteasome Endopeptidase Complex genetics, Signal Transduction drug effects, Signal Transduction genetics, Tumor Suppressor Protein p53 genetics, Ubiquitin genetics, Up-Regulation drug effects, Up-Regulation genetics, Autophagy physiology, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Tumor Suppressor Protein p53 metabolism
- Abstract
Background: Activation of wild-type p53 with the small molecule sirtuin inhibitor Tenovin-6 (Tnv-6) induces p53-dependent apoptosis in many malignant cells. In contrast, Tnv-6 reduces chronic lymphocytic leukaemia (CLL) cell viability with dysregulation of autophagy, without increasing p53-pathway activity., Methods: Here, we have investigated whether a quiescent phenotype (unique to CLL) determines the Tnv-6 response, by comparing the effects of Tnv-6 on activated and proliferating CLL. We further studied if these responses are p53-dependent., Results: Unlike quiescent cells, cell death in activated cultures treated with Tnv-6 was consistently associated with p53 upregulation. However, p53 acetylation remained unchanged, without caspase-3 cleavage or apoptosis on electron microscopy. Instead, cellular ultrastructure and protein profiles indicated autophagy inhibition, with reduced ubiquitin-proteasome activity. In specimens with mutant TP53 cultured with Tnv-6, changes in the autophagy-associated protein LC3 occurred independently of p53. Cells treated with Tnv-6 analogues lacking sirtuin inhibitory activity had attenuated LC3 lipidation compared with Tnv-6 (P0.01), suggesting that autophagy dysregulation occurs predominantly through an effect on sirtuins., Conclusion: These cell cycle and p53-independent anti-leukaemic mechanisms potentially offer novel therapeutic approaches to target leukaemia-sustaining cells in CLL, including in disease with p53-pathway dysfunction. Whether targets in addition to sirtuins contribute to autophagy dysregulation by Tnv-6, requires further investigation.
- Published
- 2013
- Full Text
- View/download PDF
50. Protein phosphatase 4 is phosphorylated and inactivated by Cdk in response to spindle toxins and interacts with γ-tubulin.
- Author
-
Voss M, Campbell K, Saranzewa N, Campbell DG, Hastie CJ, Peggie MW, Martin-Granados C, Prescott AR, and Cohen PT
- Subjects
- Amino Acid Sequence, Cell Cycle drug effects, Enzyme Activation drug effects, HEK293 Cells, HeLa Cells, Humans, Models, Biological, Molecular Sequence Data, Nocodazole pharmacology, Phosphoprotein Phosphatases chemistry, Phosphorylation drug effects, Protein Binding drug effects, Protein Interaction Mapping, Protein Kinase Inhibitors pharmacology, Protein Subunits metabolism, Spindle Apparatus drug effects, Subcellular Fractions drug effects, Subcellular Fractions metabolism, CDC2 Protein Kinase metabolism, Phosphoprotein Phosphatases metabolism, Spindle Apparatus metabolism, Tubulin metabolism
- Abstract
Many pharmaceuticals used to treat cancer target the cell cycle or mitotic spindle dynamics, such as the anti-tumor drug, paclitaxel, which stabilizes microtubules. Here we show that, in cells arrested in mitosis with the spindle toxins, nocodazole, or paclitaxel, the endogenous protein phosphatase 4 (Ppp4) complex Ppp4c-R2-R3A is phosphorylated on its regulatory (R) subunits, and its activity is inhibited. The phosphorylations are blocked by roscovitine, indicating that they may be mediated by Cdk1-cyclin B. Endogenous Ppp4c is enriched at the centrosomes in the absence and presence of paclitaxel, nocodazole, or roscovitine, and the activity of endogenous Ppp4c-R2-R3A is inhibited from G 1/S to the G 2/M phase of the cell cycle. Endogenous γ-tubulin and its associated protein, γ-tubulin complex protein 2, both of which are essential for nucleation of microtubules at centrosomes, interact with the Ppp4 complex. Recombinant γ-tubulin can be phosphorylated by Cdk1-cyclin B or Brsk1 and dephosphorylated by Ppp4c-R2-R3A in vitro. The data indicate that Ppp4c-R2-R3A regulates microtubule organization at centrosomes during cell division in response to stress signals such as spindle toxins, paclitaxel, and nocodazole, and that inhibition of the Ppp4 complex may be advantageous for treatment of some cancers.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.