49 results on '"Zeqiraj, E"'
Search Results
2. Structure of BARD1 ARD-BRCTs in complex with H2AKc15ub nucleosomes (Map1)
- Author
-
Foglizzo, M., primary, Burdett, H., additional, Wilson, M.D., additional, and Zeqiraj, E., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Cryo-EM structure of the human GS-GN complex in the inhibited state
- Author
-
Marr, L., primary and Zeqiraj, E., additional
- Published
- 2022
- Full Text
- View/download PDF
4. Regulation of canonical Wnt signalling by the ciliopathy protein MKS1 and the E2 ubiquitin-conjugating enzyme UBE2E1
- Author
-
Johnson, C, Szymanska, K, Logan, C, Adams, M, Robinson, P, Zeqiraj, E, and Wheway, G
- Abstract
Primary ciliary defects cause a group of developmental conditions known as ciliopathies. Here, we provide mechanistic insight into ciliary ubiquitin processing in cells and for mouse model lacking the ciliary protein Mks1. In vivo loss of Mks1 sensitises cells to proteasomal disruption, leading to abnormal accumulation of ubiquitinated proteins. We identified UBE2E1, an E2 ubiquitin-conjugating enzyme that polyubiquitinates β-catenin, and RNF34, an E3 ligase, as novel interactants of MKS1. UBE2E1 and MKS1 colocalised, and loss of UBE2E1 recapitulates the ciliary and Wnt signalling phenotypes observed during loss of MKS1. Levels of UBE2E1 and MKS1 are co-dependent and UBE2E1 mediates both regulatory and degradative ubiquitination of MKS1. We demonstrate that processing of phosphorylated β-catenin occurs at the ciliary base through the functional interaction between UBE2E1 and MKS1. These observations suggest that correct β-catenin levels are tightly regulated at the primary cilium by a ciliary-specific E2 (UBE2E1) and a regulatory substrate-adaptor (MKS1).
- Published
- 2022
5. Regulation of canonical Wnt signalling by ciliary protein MKS1 and Ubiquitin Proteasome System component UBE2E1
- Author
-
Szymanska, K., Boldt, K., Logan, C.V., Ueffing, M., Zeqiraj, E., Wheway, G., and Johnson, CA.
- Abstract
A functional primary cilium is a crucial cell appendage which is essential for normal, regulated signalling, and loss of the primary cilium is implicated in a suite of severe developmental conditions known as ciliopathies. The mechanisms of signal regulation by the cilium remain unclear. Previous studies have suggested links between the primary cilium/basal body, the ubiquitin proteasome system (UPS) and Wnt signalling. Here we provide further mechanistic insights into these processes in vivo by crossing the Mks1 −/− knockout mouse with the Ub G76V - GFP reporter line. We demonstrate in vivo that MKS1 is essential for normal proteasomal processing of ubiquitinated proteins, but that this only manifests as accumulation of ubiquitinated proteins when the proteasome is inhibited. We show that an increase in proteasomal enzymatic activity and Wnt signalling de-regulation in the absence of MKS1. Yeast 2-hybrid demonstrate that the UPS component UBE2E1, an E2 ubiquitin-conjugating enzyme which polyubiquitinates β-catenin, interacts with MKS1. Levels of UBE2E1 and MKS1 are co-dependent, and loss of UBE2E1 recapitulates the ciliary and Wnt signalling phenotypes observed during loss of MKS1, suggesting a functional association between the two proteins. We suggest that MKS1 regulates UBE2E1 and other UPS components at the base of the cilium, which leads to proteasomal and canonical Wnt signalling dysregulation. These findings provide further mechanistic detail of the interaction between the basal body and the UPS in regulating signal transduction through β-catenin, and confirm that the UPS plays a central role in the molecular pathogenesis of ciliopathies.
- Published
- 2020
- Full Text
- View/download PDF
6. Metabolic control of BRISC–SHMT2 assembly regulates immune signalling
- Author
-
Walden, M, Tian, L, Ross, RL, Sykora, UM, Byrne, DP, Hesketh, EL, Masandi, SK, Cassel, J, George, R, Ault, JR, El Oualid, F, Pawłowski, K, Salvino, JM, Eyers, PA, Ranson, NA, Del Galdo, F, Greenberg, RA, and Zeqiraj, E
- Abstract
Serine hydroxymethyltransferase 2 (SHMT2) regulates one-carbon transfer reactions that are essential for amino acid and nucleotide metabolism, and uses pyridoxal-5′-phosphate (PLP) as a cofactor. Apo SHMT2 exists as a dimer with unknown functions, whereas PLP binding stabilizes the active tetrameric state. SHMT2 also promotes inflammatory cytokine signalling by interacting with the deubiquitylating BRCC36 isopeptidase complex (BRISC), although it is unclear whether this function relates to metabolism. Here we present the cryo-electron microscopy structure of the human BRISC–SHMT2 complex at a resolution of 3.8 Å. BRISC is a U-shaped dimer of four subunits, and SHMT2 sterically blocks the BRCC36 active site and inhibits deubiquitylase activity. Only the inactive SHMT2 dimer—and not the active PLP-bound tetramer—binds and inhibits BRISC. Mutations in BRISC that disrupt SHMT2 binding impair type I interferon signalling in response to inflammatory stimuli. Intracellular levels of PLP regulate the interaction between BRISC and SHMT2, as well as inflammatory cytokine responses. These data reveal a mechanism in which metabolites regulate deubiquitylase activity and inflammatory signalling.
- Published
- 2019
7. Cryo-EM structure of the Human BRISC-SHMT2 complex
- Author
-
Walden, M., primary, Hesketh, E., additional, Tian, L., additional, Ranson, N.A., additional, Greenberg, R.A., additional, and Zeqiraj, E., additional
- Published
- 2019
- Full Text
- View/download PDF
8. Higher-Order Assembly of BRCC36-KIAA0157 Is Required for DUB Activity and Biological Function
- Author
-
Zeqiraj, E, Tian, L, Piggott, CA, Pillon, MC, Duffy, NM, Ceccarelli, DF, Keszei, AFA, Lorenzen, K, Kurinov, I, Orlicky, S, Gish, GD, Heck, AJR, Guarné, A, Greenberg, RA, Sicheri, F, Sub Biomol.Mass Spect. and Proteomics, Sub Biomol.Mass Spectrometry & Proteom., and Biomolecular Mass Spectrometry and Proteomics
- Subjects
Models, Molecular ,Deubiquitinating Enzymes ,Ants ,Membrane Proteins ,Cell Biology ,Crystallography, X-Ray ,Article ,Protein Structure, Secondary ,Kinetics ,HEK293 Cells ,Nuclear Matrix-Associated Proteins ,Catalytic Domain ,Animals ,Humans ,Insect Proteins ,Ubiquitin-Specific Proteases ,Protein Multimerization ,Protein Structure, Quaternary ,Molecular Biology ,HeLa Cells ,Protein Binding - Abstract
BRCC36 is a Zn2+-dependent deubiquitinating enzyme (DUB) that hydrolyzes lysine-63-linked ubiquitin chains as part of distinct macromolecular complexes that participate in either interferon signaling or DNA-damage recognition. The MPN+ domain protein BRCC36 associates with pseudo DUB MPN- proteins KIAA0157 or Abraxas, which are essential for BRCC36 enzymatic activity. To understand the basis for BRCC36 regulation, we have solved the structure of an active BRCC36-KIAA0157 heterodimer and an inactive BRCC36 homodimer. Structural and functional characterizations show how BRCC36 is switched to an active conformation by contacts with KIAA0157. Higher-order association of BRCC36 and KIAA0157 into a dimer of heterodimers (super dimers) was required for DUB activity and interaction with targeting proteins SHMT2 and RAP80. These data provide an explanation of how an inactive pseudo DUB allosterically activates a cognate DUB partner and implicates super dimerization as a new regulatory mechanism underlying BRCC36 DUB activity, subcellular localization, and biological function. We have solved structures of the active BRCC36-KIAA0157 heterodimer and inactive BRCC36-BRCC36 homodimer DUB complexes. This work reveals the basis for allosteric control of Zn2+-dependent DUB activity and biologic function through protein-protein interactions and higher-order assembly of DUB complexes.
- Published
- 2015
9. Structure of CfBRCC36-CfKIAA0157 complex (Zn Edge)
- Author
-
Zeqiraj, E., primary
- Published
- 2015
- Full Text
- View/download PDF
10. Structure of CfBRCC36-CfKIAA0157 complex (QSQ mutant)
- Author
-
Zeqiraj, E., primary
- Published
- 2015
- Full Text
- View/download PDF
11. Structure of CfBRCC36-CfKIAA0157 complex (Selenium Edge)
- Author
-
Zeqiraj, E., primary
- Published
- 2015
- Full Text
- View/download PDF
12. Structure of metal dependent enzyme DrBRCC36
- Author
-
Zeqiraj, E., primary
- Published
- 2015
- Full Text
- View/download PDF
13. Structural Basis for the Recruitment of Glycogen Synthase by Glycogenin
- Author
-
Zeqiraj, E., primary, Judd, A., additional, and Sicheri, F., additional
- Published
- 2014
- Full Text
- View/download PDF
14. Crystal Structure of RNase L in complex with 2-5A
- Author
-
Huang, H., primary, Zeqiraj, E., additional, Ceccarelli, D.F., additional, and Sicheri, F., additional
- Published
- 2014
- Full Text
- View/download PDF
15. Crystal Structure of RNase L in complex with 2-5A and AMP-PNP
- Author
-
Huang, H., primary, Zeqiraj, E., additional, Ceccarelli, D.F., additional, and Sicheri, F., additional
- Published
- 2014
- Full Text
- View/download PDF
16. Crystal structure of SspH1 LRR domain in complex PKN1 HR1b domain
- Author
-
Keszei, A.F.A., primary, Xiaojing, T., additional, Mccormick, C., additional, Zeqiraj, E., additional, Rohde, J.R., additional, Tyers, M., additional, and Sicheri, F., additional
- Published
- 2013
- Full Text
- View/download PDF
17. Crystal structure of SspH1 LRR domain
- Author
-
Keszei, A.F.A., primary, Xiaojing, T., additional, Mccormick, C., additional, Zeqiraj, E., additional, Rohde, J.R., additional, Tyers, M., additional, and Sicheri, F., additional
- Published
- 2013
- Full Text
- View/download PDF
18. TGF-beta Receptor type 1 in complex with SB431542
- Author
-
Ogunjimi, A.A., primary, Zeqiraj, E., additional, Ceccarelli, D.F., additional, and Sicheri, F., additional
- Published
- 2012
- Full Text
- View/download PDF
19. Structure of the heterotrimeric LKB1-STRADalpha-MO25alpha complex
- Author
-
Zeqiraj, E., primary and van Aalten, D.M.F., additional
- Published
- 2009
- Full Text
- View/download PDF
20. Structure of STRAD and MO25
- Author
-
Zeqiraj, E., primary, Goldie, S., additional, Alessi, D.R., additional, and van Aalten, D.M.F., additional
- Published
- 2009
- Full Text
- View/download PDF
21. Molecular glues that inhibit deubiquitylase activity and inflammatory signalling.
- Author
-
Chandler F, Reddy PAN, Bhutda S, Ross RL, Datta A, Walden M, Walker K, Di Donato S, Cassel JA, Prakesch MA, Aman A, Datti A, Campbell LJ, Foglizzo M, Bell L, Stein DN, Ault JR, Al-Awar RS, Calabrese AN, Sicheri F, Del Galdo F, Salvino JM, Greenberg RA, and Zeqiraj E
- Abstract
Deubiquitylases (DUBs) are crucial in cell signalling and are often regulated by interactions within protein complexes. The BRCC36 isopeptidase complex (BRISC) regulates inflammatory signalling by cleaving K63-linked polyubiquitin chains on Type I interferon receptors (IFNAR1). As a Zn
2+ -dependent JAMM/MPN DUB, BRCC36 is challenging to target with selective inhibitors. We discovered first-in-class inhibitors, termed BRISC molecular glues (BLUEs), which stabilise a 16-subunit BRISC dimer in an autoinhibited conformation, blocking active sites and interactions with the targeting subunit SHMT2. This unique mode of action results in selective inhibition of BRISC over related complexes with the same catalytic subunit, splice variants and other JAMM/MPN DUBs. BLUE treatment reduced interferon-stimulated gene expression in cells containing wild type BRISC, and this effect was absent when using structure-guided, inhibitor-resistant BRISC mutants. Additionally, BLUEs increase IFNAR1 ubiquitylation and decrease IFNAR1 surface levels, offering a potential new strategy to mitigate Type I interferon-mediated diseases. Our approach also provides a template for designing selective inhibitors of large protein complexes by promoting, rather than blocking, protein-protein interactions., Competing Interests: Competing interests E.Z., R.G., J.M.S., and F.S. are named co-inventors in a patent application to use BRISC inhibitors as therapeutics (WO2024115713A1). J.M.S. owns equity in Alliance Discovery, Inc and the Barer Institute, Inc, and consults for Syndeavor Therapeutics, Inc.- Published
- 2024
- Full Text
- View/download PDF
22. Systemic Sclerosis Dermal Fibroblast Exosomes Trigger Type 1 Interferon Responses in Keratinocytes via a TBK/JAK/STAT Signaling Axis.
- Author
-
Bryon J, Wasson CW, Koeppen K, Chandler F, Willis LF, Di Donato S, Klein E, Zeqiraj E, Ross RL, and Del Galdo F
- Abstract
Objective: Activation of type I interferon (IFN) response has been shown to correlate with disease activity in systemic sclerosis (SSc). It is currently unknown whether the tissue-specific type I IFN activation is a consequence of the response observed in blood or rather its source. Exosomes from SSc fibroblasts were recently shown to activate macrophages in vitro. Here, we aimed to determine the source of type I IFN signature in SSc skin biopsies and the potential role of exosomes from SSc dermal fibroblasts in the process., Methods: Skin biopsies were obtained from the forearms of healthy patients and of those with SSc and processed for dermal fibroblasts and keratinocytes. Exosomes were isolated from healthy and SSc dermal fibroblast supernatants by ultracentrifugation and added to human skin keratinocytes. Keratinocyte transcriptome was analyzed by RNA sequencing (RNA-seq) analysis. TANK-binding kinase (TBK) and JAK were inhibited using a small molecule inhibitor (GSK8612) and tofacitinib, respectively., Results: SSc skin biopsies showed the highest levels of type I IFN response in the epidermal layer. RNA-seq analysis of keratinocytes transcriptome following exposure to dermal fibroblast exosomes showed strong up-regulation of IFN signature genes induced by SSc exosomes compared to healthy control. Inhibition of TBK or JAK activity suppressed the up-regulation of the IFN signature induced by SSc exosomes., Conclusion: IFN activation of SSc keratinocytes is dependent on their crosstalk with dermal fibroblasts and inducible by extracellular exosomes. Our data indicate that SSc fibroblast exosomes contribute to the type I IFN activation in SSc skin through activation of pattern recognition receptors upstream of TBK., (© 2024 The Author(s). Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
23. The SPATA5-SPATA5L1 ATPase complex directs replisome proteostasis to ensure genome integrity.
- Author
-
Krishnamoorthy V, Foglizzo M, Dilley RL, Wu A, Datta A, Dutta P, Campbell LJ, Degtjarik O, Musgrove LJ, Calabrese AN, Zeqiraj E, and Greenberg RA
- Subjects
- Humans, Valosin Containing Protein metabolism, Valosin Containing Protein genetics, HEK293 Cells, Cell Cycle Proteins metabolism, ATPases Associated with Diverse Cellular Activities metabolism, ATPases Associated with Diverse Cellular Activities genetics, DNA Replication, Genomic Instability, Adenosine Triphosphatases metabolism, Proteostasis
- Abstract
Ubiquitin-dependent unfolding of the CMG helicase by VCP/p97 is required to terminate DNA replication. Other replisome components are not processed in the same fashion, suggesting that additional mechanisms underlie replication protein turnover. Here, we identify replisome factor interactions with a protein complex composed of AAA+ ATPases SPATA5-SPATA5L1 together with heterodimeric partners C1orf109-CINP (55LCC). An integrative structural biology approach revealed a molecular architecture of SPATA5-SPATA5L1 N-terminal domains interacting with C1orf109-CINP to form a funnel-like structure above a cylindrically shaped ATPase motor. Deficiency in the 55LCC complex elicited ubiquitin-independent proteotoxicity, replication stress, and severe chromosome instability. 55LCC showed ATPase activity that was specifically enhanced by replication fork DNA and was coupled to cysteine protease-dependent cleavage of replisome substrates in response to replication fork damage. These findings define 55LCC-mediated proteostasis as critical for replication fork progression and genome stability and provide a rationale for pathogenic variants seen in associated human neurodevelopmental disorders., Competing Interests: Declaration of interests R.A.G. is a co-founder of RADD Pharmaceuticals. None of the work in this study relates to this company., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. The UFM1 E3 ligase recognizes and releases 60S ribosomes from ER translocons.
- Author
-
Makhlouf L, Peter JJ, Magnussen HM, Thakur R, Millrine D, Minshull TC, Harrison G, Varghese J, Lamoliatte F, Foglizzo M, Macartney T, Calabrese AN, Zeqiraj E, and Kulathu Y
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Binding Sites, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Cycle Proteins ultrastructure, Cryoelectron Microscopy, Homeostasis, Intracellular Membranes metabolism, Peptidyl Transferases chemistry, Peptidyl Transferases metabolism, Peptidyl Transferases ultrastructure, Ribosomal Proteins chemistry, Ribosomal Proteins metabolism, Ribosomal Proteins ultrastructure, RNA, Transfer metabolism, SEC Translocation Channels chemistry, SEC Translocation Channels metabolism, SEC Translocation Channels ultrastructure, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins metabolism, Tumor Suppressor Proteins ultrastructure, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum ultrastructure, Protein Processing, Post-Translational, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases ultrastructure, Ribosome Subunits, Large, Eukaryotic chemistry, Ribosome Subunits, Large, Eukaryotic metabolism, Ribosome Subunits, Large, Eukaryotic ultrastructure
- Abstract
Stalled ribosomes at the endoplasmic reticulum (ER) are covalently modified with the ubiquitin-like protein UFM1 on the 60S ribosomal subunit protein RPL26 (also known as uL24)
1,2 . This modification, which is known as UFMylation, is orchestrated by the UFM1 ribosome E3 ligase (UREL) complex, comprising UFL1, UFBP1 and CDK5RAP3 (ref.3 ). However, the catalytic mechanism of UREL and the functional consequences of UFMylation are unclear. Here we present cryo-electron microscopy structures of UREL bound to 60S ribosomes, revealing the basis of its substrate specificity. UREL wraps around the 60S subunit to form a C-shaped clamp architecture that blocks the tRNA-binding sites at one end, and the peptide exit tunnel at the other. A UFL1 loop inserts into and remodels the peptidyl transferase centre. These features of UREL suggest a crucial function for UFMylation in the release and recycling of stalled or terminated ribosomes from the ER membrane. In the absence of functional UREL, 60S-SEC61 translocon complexes accumulate at the ER membrane, demonstrating that UFMylation is necessary for releasing SEC61 from 60S subunits. Notably, this release is facilitated by a functional switch of UREL from a 'writer' to a 'reader' module that recognizes its product-UFMylated 60S ribosomes. Collectively, we identify a fundamental role for UREL in dissociating 60S subunits from the SEC61 translocon and the basis for UFMylation in regulating protein homeostasis at the ER., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
25. BRCA1-BARD1 combines multiple chromatin recognition modules to bridge nascent nucleosomes.
- Author
-
Burdett H, Foglizzo M, Musgrove LJ, Kumar D, Clifford G, Campbell LJ, Heath GR, Zeqiraj E, and Wilson MD
- Subjects
- Humans, HeLa Cells, Histones metabolism, Tumor Suppressor Proteins genetics, BRCA1 Protein chemistry, BRCA1 Protein metabolism, Chromatin chemistry, Chromatin metabolism, Nucleosomes, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases metabolism
- Abstract
Chromatin association of the BRCA1-BARD1 heterodimer is critical to promote homologous recombination repair of DNA double-strand breaks (DSBs) in S/G2. How the BRCA1-BARD1 complex interacts with chromatin that contains both damage induced histone H2A ubiquitin and inhibitory H4K20 methylation is not fully understood. We characterised BRCA1-BARD1 binding and enzymatic activity to an array of mono- and di-nucleosome substrates using biochemical, structural and single molecule imaging approaches. We found that the BRCA1-BARD1 complex preferentially interacts and modifies di-nucleosomes over mono-nucleosomes, allowing integration of H2A Lys-15 ubiquitylation signals with other chromatin modifications and features. Using high speed- atomic force microscopy (HS-AFM) to monitor how the BRCA1-BARD1 complex recognises chromatin in real time, we saw a highly dynamic complex that bridges two nucleosomes and associates with the DNA linker region. Bridging is aided by multivalent cross-nucleosome interactions that enhance BRCA1-BARD1 E3 ubiquitin ligase catalytic activity. Multivalent interactions across nucleosomes explain how BRCA1-BARD1 can recognise chromatin that retains partial di-methylation at H4 Lys-20 (H4K20me2), a parental histone mark that blocks BRCA1-BARD1 interaction with nucleosomes, to promote its enzymatic and DNA repair activities., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
26. VEGFR endocytosis: Implications for angiogenesis.
- Author
-
Saikia Q, Reeve H, Alzahrani A, Critchley WR, Zeqiraj E, Divan A, Harrison MA, and Ponnambalam S
- Subjects
- Humans, Signal Transduction, Lymphangiogenesis physiology, Endocytosis, Vascular Endothelial Growth Factor A metabolism, Receptors, Vascular Endothelial Growth Factor metabolism
- Abstract
The binding of vascular endothelial growth factor (VEGF) superfamily to VEGF receptor tyrosine kinases (VEGFRs) and co-receptors regulates vasculogenesis, angiogenesis and lymphangiogenesis. A recurring theme is that dysfunction in VEGF signaling promotes pathological angiogenesis, an important feature of cancer and pro-inflammatory disease states. Endocytosis of basal (resting) or activated VEGFRs facilitates signal attenuation and endothelial quiescence. However, increasing evidence suggest that activated VEGFRs can continue to signal from intracellular compartments such as endosomes. In this chapter, we focus on the evolving link between VEGFR endocytosis, signaling and turnover and the implications for angiogenesis. There is much interest in how such understanding of VEGFR dynamics can be harnessed therapeutically for a wide range of human disease states., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
27. Autologous K63 deubiquitylation within the BRCA1-A complex licenses DNA damage recognition.
- Author
-
Jiang Q, Foglizzo M, Morozov YI, Yang X, Datta A, Tian L, Thada V, Li W, Zeqiraj E, and Greenberg RA
- Subjects
- Animals, Chromatography, Liquid, DNA Repair, HeLa Cells, Humans, Mice, Tandem Mass Spectrometry, Ubiquitin metabolism, BRCA1 Protein genetics, BRCA1 Protein metabolism, DNA Damage, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Deubiquitinating Enzymes genetics, Deubiquitinating Enzymes metabolism, Histone Chaperones genetics, Histone Chaperones metabolism
- Abstract
The BRCA1-A complex contains matching lysine-63 ubiquitin (K63-Ub) binding and deubiquitylating activities. How these functionalities are coordinated to effectively respond to DNA damage remains unknown. We generated Brcc36 deubiquitylating enzyme (DUB) inactive mice to address this gap in knowledge in a physiologic system. DUB inactivation impaired BRCA1-A complex damage localization and repair activities while causing early lethality when combined with Brca2 mutation. Damage response dysfunction in DUB-inactive cells corresponded to increased K63-Ub on RAP80 and BRCC36. Chemical cross-linking coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) and cryogenic-electron microscopy (cryo-EM) analyses of isolated BRCA1-A complexes demonstrated the RAP80 ubiquitin interaction motifs are occupied by ubiquitin exclusively in the DUB-inactive complex, linking auto-inhibition by internal K63-Ub chains to loss of damage site ubiquitin recognition. These findings identify RAP80 and BRCC36 as autologous DUB substrates in the BRCA1-A complex, thus explaining the evolution of matching ubiquitin-binding and hydrolysis activities within a single macromolecular assembly., (© 2022 Jiang et al.)
- Published
- 2022
- Full Text
- View/download PDF
28. Mechanism of glycogen synthase inactivation and interaction with glycogenin.
- Author
-
Marr L, Biswas D, Daly LA, Browning C, Vial SCM, Maskell DP, Hudson C, Bertrand JA, Pollard J, Ranson NA, Khatter H, Eyers CE, Sakamoto K, and Zeqiraj E
- Subjects
- Glucose-6-Phosphate metabolism, Glycogen metabolism, Glycoproteins metabolism, Humans, Muscle, Skeletal metabolism, Phosphorylation, Glucosyltransferases genetics, Glucosyltransferases metabolism, Glycogen Synthase genetics, Glycogen Synthase metabolism
- Abstract
Glycogen is the major glucose reserve in eukaryotes, and defects in glycogen metabolism and structure lead to disease. Glycogenesis involves interaction of glycogenin (GN) with glycogen synthase (GS), where GS is activated by glucose-6-phosphate (G6P) and inactivated by phosphorylation. We describe the 2.6 Å resolution cryo-EM structure of phosphorylated human GS revealing an autoinhibited GS tetramer flanked by two GN dimers. Phosphorylated N- and C-termini from two GS protomers converge near the G6P-binding pocket and buttress against GS regulatory helices. This keeps GS in an inactive conformation mediated by phospho-Ser641 interactions with a composite "arginine cradle". Structure-guided mutagenesis perturbing interactions with phosphorylated tails led to increased basal/unstimulated GS activity. We propose that multivalent phosphorylation supports GS autoinhibition through interactions from a dynamic "spike" region, allowing a tuneable rheostat for regulating GS activity. This work therefore provides insights into glycogen synthesis regulation and facilitates studies of glycogen-related diseases., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
29. Regulation of canonical Wnt signalling by the ciliopathy protein MKS1 and the E2 ubiquitin-conjugating enzyme UBE2E1.
- Author
-
Szymanska K, Boldt K, Logan CV, Adams M, Robinson PA, Ueffing M, Zeqiraj E, Wheway G, and Johnson CA
- Subjects
- Animals, Cilia metabolism, Humans, Mice, Mice, Knockout, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism, Ubiquitination, beta Catenin metabolism, Ciliopathies metabolism, Proteins metabolism, Ubiquitin-Conjugating Enzymes metabolism, Wnt Signaling Pathway
- Abstract
Primary ciliary defects cause a group of developmental conditions known as ciliopathies. Here, we provide mechanistic insight into ciliary ubiquitin processing in cells and for mouse model lacking the ciliary protein Mks1. In vivo loss of Mks1 sensitises cells to proteasomal disruption, leading to abnormal accumulation of ubiquitinated proteins. We identified UBE2E1, an E2 ubiquitin-conjugating enzyme that polyubiquitinates β-catenin, and RNF34, an E3 ligase, as novel interactants of MKS1. UBE2E1 and MKS1 colocalised, and loss of UBE2E1 recapitulates the ciliary and Wnt signalling phenotypes observed during loss of MKS1. Levels of UBE2E1 and MKS1 are co-dependent and UBE2E1 mediates both regulatory and degradative ubiquitination of MKS1. We demonstrate that processing of phosphorylated β-catenin occurs at the ciliary base through the functional interaction between UBE2E1 and MKS1. These observations suggest that correct β-catenin levels are tightly regulated at the primary cilium by a ciliary-specific E2 (UBE2E1) and a regulatory substrate-adaptor (MKS1)., Competing Interests: KS, KB, CL, MA, PR, MU, EZ, GW, CJ No competing interests declared, (© 2022, Szymanska et al.)
- Published
- 2022
- Full Text
- View/download PDF
30. Investigation of the specificity and mechanism of action of the ULK1/AMPK inhibitor SBI-0206965.
- Author
-
Ahwazi D, Neopane K, Markby GR, Kopietz F, Ovens AJ, Dall M, Hassing AS, Gräsle P, Alshuweishi Y, Treebak JT, Salt IP, Göransson O, Zeqiraj E, Scott JW, and Sakamoto K
- Subjects
- 3T3-L1 Cells, AMP-Activated Protein Kinases genetics, AMP-Activated Protein Kinases metabolism, Adipocytes drug effects, Adipocytes metabolism, Animals, Autophagy-Related Protein-1 Homolog genetics, Autophagy-Related Protein-1 Homolog metabolism, Benzamides metabolism, Cell Line, Cell Line, Tumor, Cells, Cultured, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Molecular Docking Simulation, Mutation, Missense, Protein Binding drug effects, Protein Multimerization, Pyrimidines metabolism, Rats, Sprague-Dawley, Recombinant Proteins chemistry, Recombinant Proteins genetics, Rats, AMP-Activated Protein Kinases antagonists & inhibitors, Autophagy-Related Protein-1 Homolog antagonists & inhibitors, Benzamides pharmacology, Pyrimidines pharmacology, Recombinant Proteins metabolism
- Abstract
SBI-0206965, originally identified as an inhibitor of the autophagy initiator kinase ULK1, has recently been reported as a more potent and selective AMP-activated protein kinase (AMPK) inhibitor relative to the widely used, but promiscuous inhibitor Compound C/Dorsomorphin. Here, we studied the effects of SBI-0206965 on AMPK signalling and metabolic readouts in multiple cell types, including hepatocytes, skeletal muscle cells and adipocytes. We observed SBI-0206965 dose dependently attenuated AMPK activator (991)-stimulated ACC phosphorylation and inhibition of lipogenesis in hepatocytes. SBI-0206965 (≥25 μM) modestly inhibited AMPK signalling in C2C12 myotubes, but also inhibited insulin signalling, insulin-mediated/AMPK-independent glucose uptake, and AICA-riboside uptake. We performed an extended screen of SBI-0206965 against a panel of 140 human protein kinases in vitro, which showed SBI-0206965 inhibits several kinases, including members of AMPK-related kinases (NUAK1, MARK3/4), equally or more potently than AMPK or ULK1. This screen, together with molecular modelling, revealed that most SBI-0206965-sensitive kinases contain a large gatekeeper residue with a preference for methionine at this position. We observed that mutation of the gatekeeper methionine to a smaller side chain amino acid (threonine) rendered AMPK and ULK1 resistant to SBI-0206965 inhibition. These results demonstrate that although SBI-0206965 has utility for delineating AMPK or ULK1 signalling and cellular functions, the compound potently inhibits several other kinases and critical cellular functions such as glucose and nucleoside uptake. Our study demonstrates a role for the gatekeeper residue as a determinant of the inhibitor sensitivity and inhibitor-resistant mutant forms could be exploited as potential controls to probe specific cellular effects of SBI-0206965., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
31. Flexible and Green Electronics Manufactured by Origami Folding of Nanosilicate-Reinforced Cellulose Paper.
- Author
-
Kadumudi FB, Trifol J, Jahanshahi M, Zsurzsan TG, Mehrali M, Zeqiraj E, Shaki H, Alehosseini M, Gundlach C, Li Q, Dong M, Akbari M, Knott A, Almdal K, and Dolatshahi-Pirouz A
- Abstract
Today's consumer electronics are made from nonrenewable and toxic components. They are also rigid, bulky, and manufactured in an energy-inefficient manner via CO
2 -generating routes. Though petroleum-based polymers such as polyethylene terephthalate and polyethylene naphthalate can address the rigidity issue, they have a large carbon footprint and generate harmful waste. Scalable routes for manufacturing electronics that are both flexible and ecofriendly (Fleco) could address the challenges in the field. Ideally, such substrates must incorporate into electronics without compromising device performance. In this work, we demonstrate that a new type of wood-based [nanocellulose (NC)] material made via nanosilicate (NS) reinforcement can yield flexible electronics that can bend and roll without loss of electrical function. Specifically, the NSs interact electrostatically with NC to reinforce thermal and mechanical properties. For instance, films containing 34 wt % of NS displayed an increased young's modulus (1.5 times), thermal stability (290 → 310 °C), and a low coefficient of thermal expansion (40 ppm/K). These films can also easily be separated and renewed into new devices through simple and low-energy processes. Moreover, we used very cheap and environmentally friendly NC from American Value Added Pulping (AVAP) technology, American Process, and therefore, the manufacturing cost of our NS-reinforced NC paper is much cheaper ($0.016 per dm-2 ) than that of conventional NC-based substrates. Looking forward, the methodology highlighted herein is highly attractive as it can unlock the secrets of Fleco electronics and transform otherwise bulky, rigid, and "difficult-to-process" rigid circuits into more aesthetic and flexible ones while simultaneously bringing relief to an already-overburdened ecosystem.- Published
- 2020
- Full Text
- View/download PDF
32. Emerging concepts in pseudoenzyme classification, evolution, and signaling.
- Author
-
Ribeiro AJM, Das S, Dawson N, Zaru R, Orchard S, Thornton JM, Orengo C, Zeqiraj E, Murphy JM, and Eyers PA
- Subjects
- Enzymes classification, Enzymes genetics, Evolution, Molecular, Signal Transduction genetics
- Abstract
The 21st century is witnessing an explosive surge in our understanding of pseudoenzyme-driven regulatory mechanisms in biology. Pseudoenzymes are proteins that have sequence homology with enzyme families but that are proven or predicted to lack enzyme activity due to mutations in otherwise conserved catalytic amino acids. The best-studied pseudoenzymes are pseudokinases, although examples from other families are emerging at a rapid rate as experimental approaches catch up with an avalanche of freely available informatics data. Kingdom-wide analysis in prokaryotes, archaea and eukaryotes reveals that between 5 and 10% of proteins that make up enzyme families are pseudoenzymes, with notable expansions and contractions seemingly associated with specific signaling niches. Pseudoenzymes can allosterically activate canonical enzymes, act as scaffolds to control assembly of signaling complexes and their localization, serve as molecular switches, or regulate signaling networks through substrate or enzyme sequestration. Molecular analysis of pseudoenzymes is rapidly advancing knowledge of how they perform noncatalytic functions and is enabling the discovery of unexpected, and previously unappreciated, functions of their intensively studied enzyme counterparts. Notably, upon further examination, some pseudoenzymes have previously unknown enzymatic activities that could not have been predicted a priori. Pseudoenzymes can be targeted and manipulated by small molecules and therefore represent new therapeutic targets (or anti-targets, where intervention should be avoided) in various diseases. In this review, which brings together broad bioinformatics and cell signaling approaches in the field, we highlight a selection of findings relevant to a contemporary understanding of pseudoenzyme-based biology., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
33. Metabolic control of BRISC-SHMT2 assembly regulates immune signalling.
- Author
-
Walden M, Tian L, Ross RL, Sykora UM, Byrne DP, Hesketh EL, Masandi SK, Cassel J, George R, Ault JR, El Oualid F, Pawłowski K, Salvino JM, Eyers PA, Ranson NA, Del Galdo F, Greenberg RA, and Zeqiraj E
- Subjects
- Cryoelectron Microscopy, Deubiquitinating Enzymes antagonists & inhibitors, Deubiquitinating Enzymes chemistry, Deubiquitinating Enzymes ultrastructure, Glycine Hydroxymethyltransferase ultrastructure, HEK293 Cells, Humans, Inflammation immunology, Models, Molecular, Multienzyme Complexes chemistry, Multienzyme Complexes genetics, Mutation, Protein Binding, Protein Multimerization, Protein Structure, Quaternary, Pyridoxal Phosphate metabolism, Deubiquitinating Enzymes metabolism, Glycine Hydroxymethyltransferase metabolism, Interferon Type I immunology, Multienzyme Complexes immunology, Multienzyme Complexes metabolism, Signal Transduction immunology
- Abstract
Serine hydroxymethyltransferase 2 (SHMT2) regulates one-carbon transfer reactions that are essential for amino acid and nucleotide metabolism, and uses pyridoxal-5'-phosphate (PLP) as a cofactor. Apo SHMT2 exists as a dimer with unknown functions, whereas PLP binding stabilizes the active tetrameric state. SHMT2 also promotes inflammatory cytokine signalling by interacting with the deubiquitylating BRCC36 isopeptidase complex (BRISC), although it is unclear whether this function relates to metabolism. Here we present the cryo-electron microscopy structure of the human BRISC-SHMT2 complex at a resolution of 3.8 Å. BRISC is a U-shaped dimer of four subunits, and SHMT2 sterically blocks the BRCC36 active site and inhibits deubiquitylase activity. Only the inactive SHMT2 dimer-and not the active PLP-bound tetramer-binds and inhibits BRISC. Mutations in BRISC that disrupt SHMT2 binding impair type I interferon signalling in response to inflammatory stimuli. Intracellular levels of PLP regulate the interaction between BRISC and SHMT2, as well as inflammatory cytokine responses. These data reveal a mechanism in which metabolites regulate deubiquitylase activity and inflammatory signalling.
- Published
- 2019
- Full Text
- View/download PDF
34. Metformin reduces liver glucose production by inhibition of fructose-1-6-bisphosphatase.
- Author
-
Hunter RW, Hughey CC, Lantier L, Sundelin EI, Peggie M, Zeqiraj E, Sicheri F, Jessen N, Wasserman DH, and Sakamoto K
- Subjects
- Adenosine Monophosphate pharmacology, Aminoimidazole Carboxamide analogs & derivatives, Aminoimidazole Carboxamide pharmacology, Animals, Base Sequence, Chickens, Disease Models, Animal, Fructose-Bisphosphatase chemistry, Fructose-Bisphosphatase genetics, Glucose Intolerance pathology, Homeostasis drug effects, Humans, Hypoglycemia pathology, Liver drug effects, Mice, Inbred C57BL, Mutation genetics, Obesity pathology, Prodrugs chemistry, Ribonucleotides pharmacology, Fructose-Bisphosphatase metabolism, Glucose biosynthesis, Liver enzymology, Metformin pharmacology
- Abstract
Metformin is a first-line drug for the treatment of individuals with type 2 diabetes, yet its precise mechanism of action remains unclear. Metformin exerts its antihyperglycemic action primarily through lowering hepatic glucose production (HGP). This suppression is thought to be mediated through inhibition of mitochondrial respiratory complex I, and thus elevation of 5'-adenosine monophosphate (AMP) levels and the activation of AMP-activated protein kinase (AMPK), though this proposition has been challenged given results in mice lacking hepatic AMPK. Here we report that the AMP-inhibited enzyme fructose-1,6-bisphosphatase-1 (FBP1), a rate-controlling enzyme in gluconeogenesis, functions as a major contributor to the therapeutic action of metformin. We identified a point mutation in FBP1 that renders it insensitive to AMP while sparing regulation by fructose-2,6-bisphosphate (F-2,6-P
2 ), and knock-in (KI) of this mutant in mice significantly reduces their response to metformin treatment. We observe this during a metformin tolerance test and in a metformin-euglycemic clamp that we have developed. The antihyperglycemic effect of metformin in high-fat diet-fed diabetic FBP1-KI mice was also significantly blunted compared to wild-type controls. Collectively, we show a new mechanism of action for metformin and provide further evidence that molecular targeting of FBP1 can have antihyperglycemic effects.- Published
- 2018
- Full Text
- View/download PDF
35. Pseudo-DUBs as allosteric activators and molecular scaffolds of protein complexes.
- Author
-
Walden M, Masandi SK, Pawłowski K, and Zeqiraj E
- Subjects
- Allosteric Regulation, Catalytic Domain, Proteasome Endopeptidase Complex metabolism, Proteins metabolism, Ubiquitin metabolism
- Abstract
The ubiquitin (Ub) proteasome system and Ub signalling networks are crucial to cell biology and disease development. Deubiquitylases (DUBs) control cell signalling by removing mono-Ub and polyubiquitin chains from substrates. DUBs take part in almost all processes that regulate cellular life and are frequently dysregulated in disease. We have catalogued 99 currently known DUBs in the human genome and sequence conservation analyses of catalytic residues suggest that 11 lack enzyme activity and are classed as pseudo-DUBs. These pseudoenzymes play important biological roles by allosterically activating catalytically competent DUBs as well as other active enzymes. Additionally, pseudoenzymes act as assembly scaffolds of macromolecular complexes. We discuss how pseudo-DUBs have lost their catalytic activity, their diverse mechanisms of action and their potential as therapeutic targets. Many known pseudo-DUBs play crucial roles in cell biology and it is likely that unstudied and overlooked pseudo-DUB genes will have equally important functions., (© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2018
- Full Text
- View/download PDF
36. Getting a handle on glycogen synthase - Its interaction with glycogenin.
- Author
-
Zeqiraj E and Sicheri F
- Subjects
- Glucose metabolism, Glycogen metabolism, Glycosylation, Humans, Glucosyltransferases metabolism, Glycogen Synthase metabolism, Glycoproteins metabolism
- Abstract
Glycogen is a polymer of glucose that serves as a major energy reserve in eukaryotes. It is synthesized through the cooperative action of glycogen synthase (GS), glycogenin (GN) and glycogen branching enzyme. GN initiates the first enzymatic step in the glycogen synthesis process by self glucosylation of a short 8-12 glucose residue primer. After interacting with GN, GS then extends this sugar primer to form glycogen particles of different sizes. We discuss recent developments in the structural biology characterization of GS and GN enzymes, which have contributed to a better understanding of how the two proteins interact and how they collaborate to synthesize glycogen particles., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
37. Higher-Order Assembly of BRCC36-KIAA0157 Is Required for DUB Activity and Biological Function.
- Author
-
Zeqiraj E, Tian L, Piggott CA, Pillon MC, Duffy NM, Ceccarelli DF, Keszei AF, Lorenzen K, Kurinov I, Orlicky S, Gish GD, Heck AJ, Guarné A, Greenberg RA, and Sicheri F
- Subjects
- Animals, Catalytic Domain, Crystallography, X-Ray, Deubiquitinating Enzymes, HEK293 Cells, HeLa Cells, Humans, Insect Proteins physiology, Kinetics, Membrane Proteins chemistry, Models, Molecular, Nuclear Matrix-Associated Proteins physiology, Protein Binding, Protein Multimerization, Protein Structure, Quaternary, Protein Structure, Secondary, Ubiquitin-Specific Proteases physiology, Ants enzymology, Insect Proteins chemistry, Nuclear Matrix-Associated Proteins chemistry, Ubiquitin-Specific Proteases chemistry
- Abstract
BRCC36 is a Zn(2+)-dependent deubiquitinating enzyme (DUB) that hydrolyzes lysine-63-linked ubiquitin chains as part of distinct macromolecular complexes that participate in either interferon signaling or DNA-damage recognition. The MPN(+) domain protein BRCC36 associates with pseudo DUB MPN(-) proteins KIAA0157 or Abraxas, which are essential for BRCC36 enzymatic activity. To understand the basis for BRCC36 regulation, we have solved the structure of an active BRCC36-KIAA0157 heterodimer and an inactive BRCC36 homodimer. Structural and functional characterizations show how BRCC36 is switched to an active conformation by contacts with KIAA0157. Higher-order association of BRCC36 and KIAA0157 into a dimer of heterodimers (super dimers) was required for DUB activity and interaction with targeting proteins SHMT2 and RAP80. These data provide an explanation of how an inactive pseudo DUB allosterically activates a cognate DUB partner and implicates super dimerization as a new regulatory mechanism underlying BRCC36 DUB activity, subcellular localization, and biological function., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
38. Expression and purification of functional human glycogen synthase-1:glycogenin-1 complex in insect cells.
- Author
-
Hunter RW, Zeqiraj E, Morrice N, Sicheri F, and Sakamoto K
- Subjects
- Animals, Humans, Recombinant Proteins biosynthesis, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Sf9 Cells, Spodoptera, Gene Expression, Glucosyltransferases biosynthesis, Glucosyltransferases genetics, Glucosyltransferases isolation & purification, Glycogen Synthase biosynthesis, Glycogen Synthase genetics, Glycogen Synthase isolation & purification, Glycoproteins biosynthesis, Glycoproteins genetics, Glycoproteins isolation & purification, Multienzyme Complexes biosynthesis, Multienzyme Complexes genetics, Multienzyme Complexes isolation & purification
- Abstract
We report the successful expression and purification of functional human muscle glycogen synthase (GYS1) in complex with human glycogenin-1 (GN1). Stoichiometric GYS1:GN1 complex was produced by co-expression of GYS1 and GN1 using a bicistronic pFastBac™-Dual expression vector, followed by affinity purification and subsequent size-exclusion chromatography. Mass spectrometry analysis identified that GYS1 is phosphorylated at several well-characterised and uncharacterised Ser/Thr residues. Biochemical analysis, including activity ratio (in the absence relative to that in the presence of glucose-6-phosphate) measurement, covalently attached phosphate estimation as well as phosphatase treatment, revealed that recombinant GYS1 is substantially more heavily phosphorylated than would be observed in intact human or rodent muscle tissues. A large quantity of highly-pure stoichiometric GYS1:GN1 complex will be useful to study its structural and biochemical properties in the future, which would reveal mechanistic insights into its functional role in glycogen biosynthesis., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
39. Structural basis for the recruitment of glycogen synthase by glycogenin.
- Author
-
Zeqiraj E, Tang X, Hunter RW, García-Rocha M, Judd A, Deak M, von Wilamowitz-Moellendorff A, Kurinov I, Guinovart JJ, Tyers M, Sakamoto K, and Sicheri F
- Subjects
- Animals, Caenorhabditis elegans genetics, Cell-Free System, Cells, Cultured, Crystallography, X-Ray, Glycogen biosynthesis, Glycogen chemistry, Glycogen genetics, Glycosylation, Mice, Mice, Knockout, Protein Binding, Protein Multimerization, Protein Structure, Quaternary, Structure-Activity Relationship, Caenorhabditis elegans enzymology, Caenorhabditis elegans Proteins chemistry, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, Glucosyltransferases chemistry, Glucosyltransferases genetics, Glucosyltransferases metabolism, Glycogen Synthase chemistry, Glycogen Synthase genetics, Glycogen Synthase metabolism, Glycoproteins chemistry, Glycoproteins genetics, Glycoproteins metabolism
- Abstract
Glycogen is a primary form of energy storage in eukaryotes that is essential for glucose homeostasis. The glycogen polymer is synthesized from glucose through the cooperative action of glycogen synthase (GS), glycogenin (GN), and glycogen branching enzyme and forms particles that range in size from 10 to 290 nm. GS is regulated by allosteric activation upon glucose-6-phosphate binding and inactivation by phosphorylation on its N- and C-terminal regulatory tails. GS alone is incapable of starting synthesis of a glycogen particle de novo, but instead it extends preexisting chains initiated by glycogenin. The molecular determinants by which GS recognizes self-glucosylated GN, the first step in glycogenesis, are unknown. We describe the crystal structure of Caenorhabditis elegans GS in complex with a minimal GS targeting sequence in GN and show that a 34-residue region of GN binds to a conserved surface on GS that is distinct from previously characterized allosteric and binding surfaces on the enzyme. The interaction identified in the GS-GN costructure is required for GS-GN interaction and for glycogen synthesis in a cell-free system and in intact cells. The interaction of full-length GS-GN proteins is enhanced by an avidity effect imparted by a dimeric state of GN and a tetrameric state of GS. Finally, the structure of the N- and C-terminal regulatory tails of GS provide a basis for understanding phosphoregulation of glycogen synthesis. These results uncover a central molecular mechanism that governs glycogen metabolism.
- Published
- 2014
- Full Text
- View/download PDF
40. Structure of an SspH1-PKN1 complex reveals the basis for host substrate recognition and mechanism of activation for a bacterial E3 ubiquitin ligase.
- Author
-
Keszei AF, Tang X, McCormick C, Zeqiraj E, Rohde JR, Tyers M, and Sicheri F
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Bacterial Proteins chemistry, Bacterial Proteins genetics, Binding Sites genetics, Catalytic Domain, Crystallography, X-Ray, HEK293 Cells, Humans, Immunoblotting, Models, Molecular, Mutation, Proteasome Endopeptidase Complex metabolism, Protein Binding, Protein Kinase C chemistry, Protein Kinase C genetics, Protein Structure, Tertiary, Receptors, Androgen metabolism, Salmonella genetics, Salmonella metabolism, Substrate Specificity, Ubiquitin-Protein Ligases genetics, Ubiquitination, Bacterial Proteins metabolism, Protein Kinase C metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
IpaH proteins are bacterium-specific E3 enzymes that function as type three secretion system (T3SS) effectors in Salmonella, Shigella, and other Gram-negative bacteria. IpaH enzymes recruit host substrates for ubiquitination via a leucine-rich repeat (LRR) domain, which can inhibit the catalytic domain in the absence of substrate. The basis for substrate recognition and the alleviation of autoinhibition upon substrate binding is unknown. Here, we report the X-ray structure of Salmonella SspH1 in complex with human PKN1. The LRR domain of SspH1 interacts specifically with the HR1b coiled-coil subdomain of PKN1 in a manner that sterically displaces the catalytic domain from the LRR domain, thereby activating catalytic function. SspH1 catalyzes the ubiquitination and proteasome-dependent degradation of PKN1 in cells, which attenuates androgen receptor responsiveness but not NF-κB activity. These regulatory features are conserved in other IpaH-substrate interactions. Our results explain the mechanism whereby substrate recognition and enzyme autoregulation are coupled in this class of bacterial ubiquitin ligases.
- Published
- 2014
- Full Text
- View/download PDF
41. Dimeric structure of pseudokinase RNase L bound to 2-5A reveals a basis for interferon-induced antiviral activity.
- Author
-
Huang H, Zeqiraj E, Dong B, Jha BK, Duffy NM, Orlicky S, Thevakumaran N, Talukdar M, Pillon MC, Ceccarelli DF, Wan LC, Juang YC, Mao DY, Gaughan C, Brinton MA, Perelygin AA, Kourinov I, Guarné A, Silverman RH, and Sicheri F
- Subjects
- Adenosine Diphosphate chemistry, Adenylyl Imidodiphosphate chemistry, Animals, Ankyrin Repeat, Binding Sites, Crystallography, X-Ray, Dimerization, Encephalomyocarditis virus, Endoribonucleases genetics, Endoribonucleases physiology, HeLa Cells, Humans, Models, Molecular, Mutagenesis, Site-Directed, Picornaviridae, Protein Structure, Tertiary, Scattering, Radiation, Structure-Activity Relationship, Sus scrofa, Adenine Nucleotides chemistry, Endoribonucleases chemistry, Oligoribonucleotides chemistry
- Abstract
RNase L is an ankyrin repeat domain-containing dual endoribonuclease-pseudokinase that is activated by unusual 2,'5'-oligoadenylate (2-5A) second messengers and which impedes viral infections in higher vertebrates. Despite its importance in interferon-regulated antiviral innate immunity, relatively little is known about its precise mechanism of action. Here we present a functional characterization of 2.5 Å and 3.25 Å X-ray crystal and small-angle X-ray scattering structures of RNase L bound to a natural 2-5A activator with and without ADP or the nonhydrolysable ATP mimetic AMP-PNP. These studies reveal how recognition of 2-5A through interactions with the ankyrin repeat domain and the pseudokinase domain, together with nucleotide binding, imposes a rigid intertwined dimer configuration that is essential for RNase catalytic and antiviral functions. The involvement of the pseudokinase domain of RNase L in 2-5A sensing, nucleotide binding, dimerization, and ribonuclease functions highlights the evolutionary adaptability of the eukaryotic protein kinase fold., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
42. A strategy for modulation of enzymes in the ubiquitin system.
- Author
-
Ernst A, Avvakumov G, Tong J, Fan Y, Zhao Y, Alberts P, Persaud A, Walker JR, Neculai AM, Neculai D, Vorobyov A, Garg P, Beatty L, Chan PK, Juang YC, Landry MC, Yeh C, Zeqiraj E, Karamboulas K, Allali-Hassani A, Vedadi M, Tyers M, Moffat J, Sicheri F, Pelletier L, Durocher D, Raught B, Rotin D, Yang J, Moran MF, Dhe-Paganon S, and Sidhu SS
- Subjects
- Amino Acid Sequence, Conserved Sequence, Drug Design, Endopeptidases chemistry, HEK293 Cells, Humans, Molecular Sequence Data, Protease Inhibitors chemistry, Protease Inhibitors pharmacology, Protein Conformation, Protein Structure, Secondary, Small Molecule Libraries, Ubiquitin chemistry, Ubiquitin genetics, Ubiquitin Thiolesterase chemistry, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases metabolism, Combinatorial Chemistry Techniques, Endopeptidases metabolism, Protease Inhibitors isolation & purification, Ubiquitin metabolism, Ubiquitin Thiolesterase metabolism, Ubiquitination drug effects
- Abstract
The ubiquitin system regulates virtually all aspects of cellular function. We report a method to target the myriad enzymes that govern ubiquitination of protein substrates. We used massively diverse combinatorial libraries of ubiquitin variants to develop inhibitors of four deubiquitinases (DUBs) and analyzed the DUB-inhibitor complexes with crystallography. We extended the selection strategy to the ubiquitin conjugating (E2) and ubiquitin ligase (E3) enzymes and found that ubiquitin variants can also enhance enzyme activity. Last, we showed that ubiquitin variants can bind selectively to ubiquitin-binding domains. Ubiquitin variants exhibit selective function in cells and thus enable orthogonal modulation of specific enzymatic steps in the ubiquitin system.
- Published
- 2013
- Full Text
- View/download PDF
43. Analysis of substrate specificity and cyclin Y binding of PCTAIRE-1 kinase.
- Author
-
Shehata SN, Hunter RW, Ohta E, Peggie MW, Lou HJ, Sicheri F, Zeqiraj E, Turk BE, and Sakamoto K
- Subjects
- Amino Acid Sequence, Binding Sites, Cyclin-Dependent Kinases chemistry, Cyclin-Dependent Kinases genetics, Cyclins genetics, HEK293 Cells, Humans, Kinetics, Mutation, Peptide Library, Protein Binding, Protein Structure, Tertiary, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Substrate Specificity, Transfection, Cyclin-Dependent Kinases metabolism, Cyclins metabolism
- Abstract
PCTAIRE-1 (cyclin-dependent kinase [CDK] 16) is a highly conserved serine/threonine kinase that belongs to the CDK family of protein kinases. Little is known regarding PCTAIRE-1 regulation and function and no robust assay exists to assess PCTAIRE-1 activity mainly due to a lack of information regarding its preferred consensus motif and the lack of bona fide substrates. We used positional scanning peptide library technology and identified the substrate-specificity requirements of PCTAIRE-1 and subsequently elaborated a peptide substrate termed PCTAIRE-tide. Recombinant PCTAIRE-1 displayed vastly improved enzyme kinetics on PCTAIRE-tide compared to a widely used generic CDK substrate peptide. PCTAIRE-tide also greatly improved detection of endogenous PCTAIRE-1 activity. Similar to other CDKs, PCTAIRE-1 requires a proline residue immediately C-terminal to the phosphoacceptor site (+1) for optimal activity. PCTAIRE-1 has a unique preference for a basic residue at +4, but not at +3 position (a key characteristic for CDKs). We also demonstrate that PCTAIRE-1 binds to a novel cyclin family member, cyclin Y, which increased PCTAIRE-1 activity towards PCTAIRE-tide >100-fold. We hypothesised that cyclin Y binds and activates PCTAIRE-1 in a way similar to which cyclin A2 binds and activates CDK2. Point mutants of cyclin Y predicted to disrupt PCTAIRE-1-cyclin Y binding severely prevented complex formation and activation of PCTAIRE-1. We have identified PCTAIRE-tide as a powerful tool to study the regulation of PCTAIRE-1. Our understanding of the molecular interaction between PCTAIRE-1 and cyclin Y further facilitates future investigation of the functions of PCTAIRE-1 kinase., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
44. Structural basis for specificity of TGFβ family receptor small molecule inhibitors.
- Author
-
Ogunjimi AA, Zeqiraj E, Ceccarelli DF, Sicheri F, Wrana JL, and David L
- Subjects
- Activin Receptors, Type I antagonists & inhibitors, Activin Receptors, Type I chemistry, Activin Receptors, Type I genetics, Activins metabolism, Benzamides chemistry, Benzamides metabolism, Bone Morphogenetic Proteins chemistry, Bone Morphogenetic Proteins genetics, Crystallography, X-Ray, Dioxoles chemistry, Dioxoles metabolism, Drug Design, HEK293 Cells, Humans, Inhibitory Concentration 50, Models, Molecular, Mutation, Phosphorylation, Plasmids, Protein Binding, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta antagonists & inhibitors, Receptors, Transforming Growth Factor beta chemistry, Receptors, Transforming Growth Factor beta genetics, Serine genetics, Substrate Specificity, Transfection, Transforming Growth Factor beta metabolism, Activin Receptors, Type I metabolism, Benzamides pharmacology, Bone Morphogenetic Proteins metabolism, Dioxoles pharmacology, Protein Serine-Threonine Kinases metabolism, Receptors, Transforming Growth Factor beta metabolism, Serine metabolism, Signal Transduction
- Abstract
Transforming growth factor-β (TGFβ) receptor kinase inhibitors have a great therapeutic potential. SB431542 is one of the mainly used kinase inhibitors of the TGFβ/Activin pathway receptors, but needs improvement of its EC(50) (EC(50)=1 μM) to be translated to clinical use. A key feature of SB431542 is that it specifically targets receptors from the TGFβ/Activin pathway but not the closely related receptors from the bone morphogenic proteins (BMP) pathway. To understand the mechanisms of this selectivity, we solved the crystal structure of the TGFβ type I receptor (TβRI) kinase domain in complex with SB431542. We mutated TβRI residues coordinating SB431542 to their counterparts in activin-receptor like kinase 2 (ALK2), a BMP receptor kinase, and tested the kinase activity of mutated TβRI. We discovered that a Ser280Thr mutation yielded a TβRI variant that was resistant to SB431542 inhibition. Furthermore, the corresponding Thr283Ser mutation in ALK2 yielded a BMP receptor sensitive to SB431542. This demonstrated that Ser280 is the key determinant of selectivity for SB431542. This work provides a framework for optimising the SB431542 scaffold to more potent and selective inhibitors of the TGFβ/Activin pathway., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
45. MO25 is a master regulator of SPAK/OSR1 and MST3/MST4/YSK1 protein kinases.
- Author
-
Filippi BM, de los Heros P, Mehellou Y, Navratilova I, Gourlay R, Deak M, Plater L, Toth R, Zeqiraj E, and Alessi DR
- Subjects
- Animals, Enzyme Activation physiology, Escherichia coli, HEK293 Cells, Humans, Immunoblotting, Insecta, Phosphorylation, Protein Isoforms metabolism, RNA, Small Interfering genetics, Calcium-Binding Proteins metabolism, Homeostasis physiology, Intracellular Signaling Peptides and Proteins metabolism, Morphogenesis physiology, Protein Serine-Threonine Kinases metabolism
- Abstract
Mouse protein-25 (MO25) isoforms bind to the STRAD pseudokinase and stabilise it in a conformation that can activate the LKB1 tumour suppressor kinase. We demonstrate that by binding to several STE20 family kinases, MO25 has roles beyond controlling LKB1. These new MO25 targets are SPAK/OSR1 kinases, regulators of ion homeostasis and blood pressure, and MST3/MST4/YSK1, involved in controlling development and morphogenesis. Our analyses suggest that MO25α and MO25β associate with these STE20 kinases in a similar manner to STRAD. MO25 isoforms induce approximately 100-fold activation of SPAK/OSR1 dramatically enhancing their ability to phosphorylate the ion cotransporters NKCC1, NKCC2 and NCC, leading to the identification of several new phosphorylation sites. siRNA-mediated reduction of expression of MO25 isoforms in mammalian cells inhibited phosphorylation of endogenous NKCC1 at residues phosphorylated by SPAK/OSR1, which is rescued by re-expression of MO25α. MO25α/β binding to MST3/MST4/YSK1 also stimulated kinase activity three- to four-fold. MO25 has evolved as a key regulator of a group of STE20 kinases and may represent an ancestral mechanism of regulating conformation of pseudokinases and activating catalytically competent protein kinases.
- Published
- 2011
- Full Text
- View/download PDF
46. Pseudokinases-remnants of evolution or key allosteric regulators?
- Author
-
Zeqiraj E and van Aalten DM
- Subjects
- Allosteric Regulation, Amino Acid Sequence, Animals, Humans, Molecular Sequence Data, Nucleotides metabolism, Protein Kinases chemistry, Protein Kinases genetics, Pseudogenes, Evolution, Molecular, Protein Kinases metabolism
- Abstract
Protein kinases provide a platform for the integration of signal transduction networks. A key feature of transmitting these cellular signals is the ability of protein kinases to activate one another by phosphorylation. A number of kinases are predicted by sequence homology to be incapable of phosphoryl group transfer due to degradation of their catalytic motifs. These are termed pseudokinases and because of the assumed lack of phosphoryltransfer activity their biological role in cellular transduction has been mysterious. Recent structure-function studies have uncovered the molecular determinants for protein kinase inactivity and have shed light to the biological functions and evolution of this enigmatic subset of the human kinome. Pseudokinases act as signal transducers by bringing together components of signalling networks, as well as allosteric activators of active protein kinases., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
47. Structure of the LKB1-STRAD-MO25 complex reveals an allosteric mechanism of kinase activation.
- Author
-
Zeqiraj E, Filippi BM, Deak M, Alessi DR, and van Aalten DM
- Subjects
- AMP-Activated Protein Kinase Kinases, AMP-Activated Protein Kinases metabolism, Adaptor Proteins, Vesicular Transport metabolism, Allosteric Regulation, Amino Acid Sequence, Binding Sites, Calcium-Binding Proteins metabolism, Crystallography, X-Ray, Enzyme Activation, Humans, Models, Molecular, Molecular Sequence Data, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism, Mutant Proteins chemistry, Mutant Proteins metabolism, Mutation, Phosphorylation, Protein Binding, Protein Conformation, Protein Interaction Domains and Motifs, Protein Serine-Threonine Kinases metabolism, Protein Structure, Tertiary, Adaptor Proteins, Vesicular Transport chemistry, Calcium-Binding Proteins chemistry, Protein Serine-Threonine Kinases chemistry
- Abstract
The LKB1 tumor suppressor is a protein kinase that controls the activity of adenosine monophosphate-activated protein kinase (AMPK). LKB1 activity is regulated by the pseudokinase STRADalpha and the scaffolding protein MO25alpha through an unknown, phosphorylation-independent, mechanism. We describe the structure of the core heterotrimeric LKB1-STRADalpha-MO25alpha complex, revealing an unusual allosteric mechanism of LKB1 activation. STRADalpha adopts a closed conformation typical of active protein kinases and binds LKB1 as a pseudosubstrate. STRADalpha and MO25alpha promote the active conformation of LKB1, which is stabilized by MO25alpha interacting with the LKB1 activation loop. This previously undescribed mechanism of kinase activation may be relevant to understanding the evolution of other pseudokinases. The structure also reveals how mutations found in Peutz-Jeghers syndrome and in various sporadic cancers impair LKB1 function.
- Published
- 2009
- Full Text
- View/download PDF
48. ATP and MO25alpha regulate the conformational state of the STRADalpha pseudokinase and activation of the LKB1 tumour suppressor.
- Author
-
Zeqiraj E, Filippi BM, Goldie S, Navratilova I, Boudeau J, Deak M, Alessi DR, and van Aalten DM
- Subjects
- AMP-Activated Protein Kinase Kinases, Abnormalities, Multiple enzymology, Adenosine Diphosphate metabolism, Amino Acid Sequence, Binding Sites, Cell Line, Conserved Sequence, Cyclin-Dependent Kinases metabolism, Cyclins metabolism, Enzyme Activation, Enzyme Stability, Humans, Magnesium, Models, Biological, Models, Molecular, Molecular Sequence Data, Mutation genetics, Protein Binding, Protein Structure, Secondary, Surface Properties, Syndrome, Adaptor Proteins, Vesicular Transport chemistry, Adaptor Proteins, Vesicular Transport metabolism, Adenosine Triphosphate metabolism, Calcium-Binding Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Pseudokinases lack essential residues for kinase activity, yet are emerging as important regulators of signal transduction networks. The pseudokinase STRAD activates the LKB1 tumour suppressor by forming a heterotrimeric complex with LKB1 and the scaffolding protein MO25. Here, we describe the structure of STRADalpha in complex with MO25alpha. The structure reveals an intricate web of interactions between STRADalpha and MO25alpha involving the alphaC-helix of STRADalpha, reminiscent of the mechanism by which CDK2 interacts with cyclin A. Surprisingly, STRADalpha binds ATP and displays a closed conformation and an ordered activation loop, typical of active protein kinases. Inactivity is accounted for by nonconservative substitution of almost all essential catalytic residues. We demonstrate that binding of ATP enhances the affinity of STRADalpha for MO25alpha, and conversely, binding of MO25alpha promotes interaction of STRADalpha with ATP. Mutagenesis studies reveal that association of STRADalpha with either ATP or MO25alpha is essential for LKB1 activation. We conclude that ATP and MO25alpha cooperate to maintain STRADalpha in an "active" closed conformation required for LKB1 activation. It has recently been demonstrated that a mutation in human STRADalpha that truncates a C-terminal region of the pseudokinase domain leads to the polyhydramnios, megalencephaly, symptomatic epilepsy (PMSE) syndrome. We demonstrate this mutation destabilizes STRADalpha and prevents association with LKB1. In summary, our findings describe one of the first structures of a genuinely inactive pseudokinase. The ability of STRADalpha to activate LKB1 is dependent on a closed "active" conformation, aided by ATP and MO25alpha binding. Thus, the function of STRADalpha is mediated through an active kinase conformation rather than kinase activity. It is possible that other pseudokinases exert their function through nucleotide binding and active conformations., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2009
- Full Text
- View/download PDF
49. Proteome analysis of metastatic colorectal cancer cells recognized by the lectin Helix pomatia agglutinin (HPA).
- Author
-
Saint-Guirons J, Zeqiraj E, Schumacher U, Greenwell P, and Dwek M
- Subjects
- Cell Line, Tumor, Electrophoresis, Polyacrylamide Gel, Humans, Lectins drug effects, Lectins immunology, Membrane Proteins immunology, Microscopy, Confocal, N-Acetylgalactosaminyltransferases pharmacology, N-Acetylglucosaminyltransferases pharmacology, N-Acetylneuraminic Acid pharmacology, Prognosis, Protein Binding, Sensitivity and Specificity, Structure-Activity Relationship, Tumor Cells, Cultured, Colorectal Neoplasms diagnosis, Colorectal Neoplasms secondary, Lectins analysis, Proteomics
- Abstract
The lectin from Helix pomatia (HPA) binds to adenocarcinomas with a metastatic phenotype but the glycoconjugates of cancer cells that bind to the lectin have yet to be characterized in detail. We used a model of metastatic (HT29) and nonmetastatic (SW480) human colorectal cancer cells and a proteomic approach to identify HPA binding glycoproteins. Cell membrane proteins purified by HPA affinity chromatography, were separated by 2-DE and analyzed by MS. Competitive inhibition experiments with N-acetylgalactosamine, N-acetylglucosamine, and sialic acid confirmed that HPA binding was via a glycan-mediated interaction. Western blot analysis showed that HPA binds to proteins not recognized by an antibody against blood group A epitope. The proteomic study showed the main HPA binding partners include integrin alphav/alpha6 and annexin A2/A4. These proteins were found complexed with microfilament proteins alpha and beta tubulin, actin, and cytokeratins 8 and 18. HPA also bound to Hsp70, Hsp90, TRAP-1, and tumor rejection factor 1. This study revealed that the prognostic utility of HPA lies in its ability to bind simultaneously to many glycoproteins involved in cell migration and signaling, in addition, the proteins recognized by HPA are glycosylated with structures distinct from the blood group A epitope.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.