37 results on '"Pseudokinase"'
Search Results
2. The expanding landscape of canonical and non-canonical protein phosphorylation.
- Author
-
Houles, Thibault, Yoon, Sang-Oh, and Roux, Philippe P.
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *PROTEIN kinases , *CELL communication , *MOLECULAR switches , *CATALYTIC activity - Abstract
The phosphoproteome is vast and intricate, with many aspects of its extent and site-specific functional roles still largely unexplored. While traditional studies have primarily focused on Ser/Thr and Tyr phosphorylation, new methodologies are emerging to study non-canonical phosphorylation events. Protein phosphatases play critical roles in cellular signaling, with recent studies highlighting their complexity and involvement in various biological processes. A significant portion of the kinome, referred to as the 'dark' kinome, remains understudied and holds great potential for novel discoveries. Pseudokinases and pseudophosphatases, despite lacking catalytic activity, play crucial roles in cell signaling. Recent advances in experimental and computational analyses have improved the understanding of kinase–substrate and phosphatase–substrate interactions. Protein phosphorylation is a crucial regulatory mechanism in cell signaling, acting as a molecular switch that modulates protein function. Catalyzed by protein kinases and reversed by phosphoprotein phosphatases, it is essential in both normal physiological and pathological states. Recent advances have uncovered a vast and intricate landscape of protein phosphorylation that include histidine phosphorylation and more unconventional events, such as pyrophosphorylation and polyphosphorylation. Many questions remain about the true size of the phosphoproteome and, more importantly, its site-specific functional relevance. The involvement of unconventional actors such as pseudokinases and pseudophosphatases adds further complexity to be resolved. This review explores recent discoveries and ongoing challenges, highlighting the need for continued research to fully elucidate the roles and regulation of protein phosphorylation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Looking lively: emerging principles of pseudokinase signaling.
- Author
-
Sheetz, Joshua B. and Lemmon, Mark A.
- Subjects
- *
PROTEIN kinases , *SMALL molecules , *PROTEIN folding , *DRUG target , *G proteins , *G protein coupled receptors - Abstract
Progress towards understanding catalytically 'dead' protein kinases – pseudokinases – in biology and disease has hastened over the past decade. An especially lively area for structural biology, pseudokinases appear to be strikingly similar to their kinase relatives, despite lacking key catalytic residues. Distinct active- and inactive-like conformation states, which are crucial for regulating bona fide protein kinases, are conserved in pseudokinases and appear to be essential for function. We discuss recent structural data on conformational transitions and nucleotide binding by pseudokinases, from which some common principles emerge. In both pseudokinases and bona fide kinases, a conformational toggle appears to control the ability to interact with signaling effectors. We also discuss how biasing this conformational toggle may provide opportunities to target pseudokinases pharmacologically in disease. Approximately 10% of proteins (58) in the human kinome are pseudokinases: they display the protein kinase fold but lack key conserved residues or have been observed experimentally to lack protein kinase activity. Several pseudokinases are implicated in human diseases, making them potential drug targets. A few domains first thought to qualify as pseudokinases retain kinase activity through compensatory changes, or have alternative catalytic activities. Many pseudokinases have compensatory mutations that allow retained ATP binding, despite being inactive kinases. Like their bona fide kinase relatives, several pseudokinases toggle conformationally between active-like and inactive-like states, and this controls their ability to interact with signaling effectors – suggesting analogies to regulation by small G proteins. Insights into conformation-dependent signaling by pseudokinases set the stage for their pharmacological targeting with conformational disruptors, and one pseudokinase-binding small molecule (targeting TYK2) is now in clinical development. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
4. Pseudokinases repurpose flexibility signatures associated with the protein kinase fold for noncatalytic roles.
- Author
-
Paul, Anindita, Subhadarshini, Seemadri, and Srinivasan, Narayanaswamy
- Abstract
The bilobal protein kinase‐like fold in pseudokinases lack one or more catalytic residues, conserved in canonical protein kinases, and are considered enzymatically deficient. Tertiary structures of pseudokinases reveal that their loops topologically equivalent to activation segments of kinases adopt contracted configurations, which is typically extended in active conformation of kinases. Herein, anisotropic network model based normal mode analysis (NMA) was conducted on 51 active conformation structures of protein kinases and 26 crystal structures of pseudokinases. Our observations indicate that although backbone fluctuation profiles are similar for individual kinase‐pseudokinase families, low intensity mean square fluctuations in pseudo‐activation segment and other sub‐structures impart rigidity to pseudokinases. Analyses of collective motions from functional modes reveal that pseudokinases, compared to active kinases, undergo distinct conformational transitions using the same structural fold. All‐atom NMA of protein kinase‐pseudokinase pairs from each family, sharing high amino acid sequence identities, yielded distinct community clusters, partitioned by residues exhibiting highly correlated fluctuations. It appears that atomic fluctuations from equivalent activation segments guide community membership and network topologies for respective kinase and pseudokinase. Our findings indicate that such adaptations in backbone and side‐chain fluctuations render pseudokinases competent for catalysis‐independent roles. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. Divergent kinase regulates membrane ultrastructure of the Toxoplasma parasitophorous vacuole.
- Author
-
Beraki, Tsebaot, Xiaoyu Hu, Broncel, Malgorzata, Young, Joanna C., O'Shaughnessy, William J., Borek, Dominika, Treeck, Moritz, and Reese, Michael L.
- Subjects
- *
TOXOPLASMA gondii , *KINASE regulation , *BIOLOGICAL membranes , *MICROBIAL virulence , *ORIGIN of life - Abstract
Apicomplexan parasites replicate within a protective organelle, called the parasitophorous vacuole (PV). The Toxoplasma gondii PV is filled with a network of tubulated membranes, which are thought to facilitate trafficking of effectors and nutrients. Despite being critical to parasite virulence, there is scant mechanistic understanding of the network's functions. Here, we identify the parasite-secreted kinase WNG1 (With-No-Gly-loop) as a critical regulator of tubular membrane biogenesis. WNG1 family members adopt an atypical protein kinase fold lacking the glycine rich ATP-binding loop that is required for catalysis in canonical kinases. Unexpectedly, we find that WNG1 is an active protein kinase that localizes to the PV lumen and phosphorylates PV-resident proteins, several of which are essential for the formation of a functional intravacuolar network. Moreover, we show that WNG1-dependent phosphorylation of these proteins is required for their membrane association, and thus their ability to tubulate membranes. Consequently, WNG1 knockout parasites have an aberrant PV membrane ultrastructure. Collectively, our results describe a unique family of Toxoplasma kinases and implicate phosphorylation of secreted proteins as a mechanism of regulating PV development during parasite infection. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
6. Functional characterization of PEAK3/C19orf35 pseudokinase and its role in regulation of CrkII-dependent signaling
- Author
-
Lopez, Mitchell
- Subjects
Biochemistry ,Biology ,CrkII ,kinase ,Motility ,PEAK3 ,Pseudokinase - Abstract
Members of the New Kinase Family 3 (NKF3), PEAK1/SgK269 and Pragmin/SgK223 pseudokinases, have emerged as important regulators of cell motility and cancer progression. Here, we demonstrate for the first time that C19orf35 (PEAK3), a newly identified member of the NKF3 family, is a kinase-like protein evolutionarily conserved across mammals and birds and a novel regulator of cell motility. In contrast to its family members, which promote cell elongation when overexpressed in cells, PEAK3 overexpression does not have an elongating effect on cell shape but instead is associated with loss of actin filaments. Through an unbiased search for PEAK3 binding partners, we identified several regulators of cell motility, including the adaptor protein CrkII. We show that by binding to CrkII, PEAK3 prevents the formation of CrkII-dependent membrane ruffling. This function of PEAK3 is reliant upon its dimerization, which is mediated through a split helical dimerization (SHED) domain conserved among all NKF3 family members. Disruption of the conserved DFG motif in the PEAK3 pseudokinase domain also interferes with its ability to dimerize and subsequently bind CrkII, suggesting that the conformation of the pseudokinase domain might play an important role in PEAK3 signaling. Hence, our data identify PEAK3 as an NKF3 family member with a unique role in cell motility driven by dimerization of its pseudokinase domain.
- Published
- 2019
7. Characterization of the cellular signaling mechanisms of Tribbles pseudokinases
- Author
-
Kung, Jennifer Elaine
- Subjects
Biochemistry ,COP1 ,kinase ,nuclear export ,pseudokinase ,tribbles ,ubiquitin ligase - Abstract
Pseudokinases are members of the protein kinase superfamily that are predicted to be catalytically inactive despite retaining the conserved kinase domain fold. Consequently, pseudokinases have evolved to signal exclusively through non-catalytic mechanisms, such as allostery and scaffolding. Though pseudokinases have been implicated in a wide range of diseases and are known to be involved in critical cellular processes, their lack of catalytic activity has made their functions challenging to study. Members of the Tribbles family of pseudokinases have recently emerged as key regulators of diverse signaling pathways, including the MAPK cascade, proteasome-dependent degradation, the TGF-β/BMP pathway, and the PI3K/Akt pathway. However, the molecular mechanisms underlying Tribbles-mediated signaling are poorly understood. One of the most highly conserved functions of Tribbles pseudokinases is their role in promoting ubiquitination of C/EBP transcription factors by the E3 ubiquitin ligase COP1. Dysregulation of this function has been implicated in acute myeloid leukemia, where overexpression of Trb1 or Trb2 is sufficient to induce leukemogenesis in mice through depletion of C/EBPα. Tribbles pseudokinases are thought to function as scaffolds to facilitate interactions between COP1 and substrates like C/EBPα, but the molecular basis for this function is only beginning to be uncovered. Here, we have investigated the molecular and structural determinants of regulation of COP1 by the pseudokinase Trb1. In doing so, we have uncovered a previously uncharacterized role for Trb1 in promoting COP1 nuclear localization through disrupting a conserved intramolecular regulatory interaction within COP1, and we have identified key structural elements that control COP1 localization. Collectively, these findings will deepen our understanding of the mechanisms underlying regulation of COP1 by Tribbles pseudokinases and broaden our knowledge of the diverse signaling functions of the Tribbles family.
- Published
- 2018
8. Cataloguing the dead: breathing new life into pseudokinase research
- Author
-
Natarajan Kannan, Dominic P. Byrne, Patrick A. Eyers, John A. Harris, and Safal Shrestha
- Subjects
0301 basic medicine ,Cell signaling ,ephrin receptor ,sequence analysis ,Sequence analysis ,pseudokinase ,Computational biology ,Review Article ,Protein Serine-Threonine Kinases ,Biochemistry ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,PSKH1 ,PSKH2 ,Humans ,Kinome ,signalling ,Protein kinase A ,Molecular Biology ,Review Articles ,Receptors, Eph Family ,biology ,Kinase ,Erythropoietin-producing hepatocellular (Eph) receptor ,tribbles ,Cell Biology ,bioinformatics ,inhibitor ,030104 developmental biology ,Histone ,030220 oncology & carcinogenesis ,pseudoenzyme ,biology.protein ,Eph tyrosine kinase ,Signal Transduction - Abstract
Pseudoenzymes are present within many, but not all, known enzyme families and lack one or more conserved canonical amino acids that help define their catalytically active counterparts. Recent findings in the pseudokinase field confirm that evolutionary repurposing of the structurally defined bilobal protein kinase fold permits distinct biological functions to emerge, many of which rely on conformational switching, as opposed to canonical catalysis. In this analysis, we evaluate progress in evaluating several members of the ‘dark’ pseudokinome that are pertinent to help drive this expanding field. Initially, we discuss how adaptions in erythropoietin‐producing hepatocellular carcinoma (Eph) receptor tyrosine kinase domains resulted in two vertebrate pseudokinases, EphA10 and EphB6, in which co‐evolving sequences generate new motifs that are likely to be important for both nucleotide binding and catalysis‐independent signalling. Secondly, we discuss how conformationally flexible Tribbles pseudokinases, which have radiated in the complex vertebrates, control fundamental aspects of cell signalling that may be targetable with covalent small molecules. Finally, we show how species‐level adaptions in the duplicated canonical kinase protein serine kinase histone (PSKH)1 sequence have led to the appearance of the pseudokinase PSKH2, whose physiological role remains mysterious. In conclusion, we show how the patterns we discover are selectively conserved within specific pseudokinases, and that when they are modelled alongside closely related canonical kinases, many are found to be located in functionally important regions of the conserved kinase fold. Interrogation of these patterns will be useful for future evaluation of these, and other, members of the unstudied human kinome., Here, we evaluate several families of poorly studied 'dark' kinases and pseudokinases using a bioinformatic and structural analysis approach. Our investigation revealed several new features of two erythropoietin‐producing hepatocellular carinoma (Eph) psedudokinases: EphA10 and EphB6. Furthermore, we discuss newly identified features of the Tribbles pseudokinases as well as the unique pseudokinase protein serine kinase histone 2 (PSKH2).
- Published
- 2020
9. KinCon: Cell‐based recording of full‐length kinase conformations
- Author
-
Eduard Stefan, Philipp Tschaikner, Florian Enzler, and Rainer Schneider
- Subjects
AMPK ,0301 basic medicine ,MAPK/ERK pathway ,Protein Conformation ,Clinical Biochemistry ,Biosensing Techniques ,Biochemistry ,Phosphotransferase ,0302 clinical medicine ,Genes, Reporter ,PKA ,Luciferases ,scaffolding function ,Kinase ,Drug discovery ,Chemistry ,kinase dimer ,Hypothesis ,CRAF ,Small molecule ,MEK ,030220 oncology & carcinogenesis ,STRADα ,kinase drug efficacies ,kinase conformations ,LKB1 ,oncokinase ,Allosteric regulation ,pseudokinase ,Computational biology ,biosensor ,BRAF ,03 medical and health sciences ,Genetics ,Humans ,Molecular Biology ,undruggable ,allosteric inhibitor ,KSR ,drug side effect ,RAF ,Cell Biology ,MAPK ,030104 developmental biology ,Luminescent Measurements ,molecular interaction ,pseudoenzyme ,Protein abundance ,Protein Kinases ,RAS ,Cell based - Abstract
The spectrum of kinase alterations displays distinct functional characteristics and requires kinase mutation‐oriented strategies for therapeutic interference. Besides phosphotransferase activity, protein abundance, and intermolecular interactions, particular patient‐mutations promote pathological kinase conformations. Despite major advances in identifying lead molecules targeting clinically relevant oncokinase functions, still many kinases are neglected and not part of drug discovery efforts. One explanation is attributed to challenges in tracking kinase activities. Chemical probes are needed to functionally annotate kinase functions, whose activities may not always depend on catalyzing phospho‐transfer. Such non‐catalytic kinase functions are related to transitions of full‐length kinase conformations. Recent findings underline that cell‐based reporter systems can be adapted to record conformation changes of kinases. Here, we discuss the possible applications of an extendable kinase conformation (KinCon) reporter toolbox for live‐cell recording of kinase states. KinCon is a genetically encoded bioluminescence‐based biosensor platform, which can be subjected for measurements of conformation dynamics of mutated kinases upon small molecule inhibitor exposure. We hypothesize that such biosensors can be utilized to delineate the molecular modus operandi for kinase and pseudokinase regulation. This should pave the path for full‐length kinase‐targeted drug discovery efforts aiming to identify single and combinatory kinase inhibitor therapies with increased specificity and efficacy.
- Published
- 2020
- Full Text
- View/download PDF
10. Molecular Mechanism of CCAAT-Enhancer Binding Protein Recruitment by the TRIB1 Pseudokinase.
- Author
-
Murphy, James M., Nakatani, Yoshio, Jamieson, Sam A., Dai, Weiwen, Lucet, Isabelle S., and Mace, Peter D.
- Subjects
- *
PROTEIN binding , *CARRIER proteins , *BIOPHYSICS , *LIGASES , *ENZYMES - Abstract
Summary CCAAT-enhancer binding proteins (C/EBPs) are transcription factors that play a central role in the differentiation of myeloid cells and adipocytes. Tribbles pseudokinases govern levels of C/EBPs by recruiting them to the COP1 ubiquitin ligase for ubiquitination. Here, we present the first crystal structure of a Tribbles protein, which reveals a catalytically inactive TRIB1 pseudokinase domain with a unique adaptation in the αC helix. A second crystal structure and biophysical studies of TRIB1 with its C-terminal extension, which includes the COP1-binding motif, show that the C-terminal extension is sequestered at a site formed by the modified TRIB1 αC helix. In addition, we have identified and characterized the TRIB1 substrate-recognition sequence within C/EBPα, which is evolutionarily conserved in C/EBP transcription factors. Binding studies indicate that C/EBPα recruitment is weaker in the presence of the C-terminal COP1-binding motif, but the magnitude of this effect suggests that the two bind distinct rather directly overlapping binding sites. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
11. The Tribbles 2 (TRB2) pseudokinase binds to ATP and autophosphorylates in a metal-independent manner.
- Author
-
Bailey, Fiona P., Byrne, Dominic P., Oruganty, Krishnadev, Eyers, Claire E., Novotny, Christopher J., Shokat, Kevan M., Kannan, Natarajan, and Eyers, Patrick A.
- Subjects
- *
CHEMICAL biology , *CALCIUM-dependent protein kinase , *GENETIC overexpression , *AUTOPHOSPHORYLATION , *ADENOSINE triphosphate , *THERMAL stability , *TARGETED drug delivery , *CELLULAR signal transduction - Abstract
The human Tribbles (TRB)-related pseudokinases are CAMK (calcium/calmodulin-dependent protein kinase)-related family members that have evolved a series of highly unusual motifs in the 'pseudocatalytic' domain. In canonical kinases, conserved amino acids bind to divalent metal ions and align ATP prior to efficient phosphoryl-transfer to substrates. However, in pseudokinases, atypical residues give rise to diverse and often unstudied biochemical and structural features that are thought to be central to cellular functions. TRB proteins play a crucial role in multiple signalling networks and overexpression confers cancer phenotypes on human cells, marking TRB pseudokinases out as a novel class of drug target. In the present paper, we report that the human pseudokinase TRB2 retains the ability to both bind and hydrolyse ATP weakly in vitro. Kinase activity is metalindependent and involves a catalytic lysine residue, which is conserved in TRB proteins throughout evolution alongside several unique amino acids in the active site. A similar low level of autophosphorylation is also preserved in the closely related human TRB3. By employing chemical genetics, we establish that the nucleotide-binding site of an 'analogue-sensitive' (AS) TRB2 mutant can be targeted with specific bulky ligands of the pyrazolopyrimidine (PP) chemotype. Our analysis confirms that TRB2 retains low levels of ATP binding and/or catalysis that is targetable with small molecules. Given the significant clinical successes associated with targeting of cancer-associated kinases with small molecule inhibitors, it is likely that similar approaches will be useful for further evaluating the TRB pseudokinases, with the translation of this information likely to furnish new leads for drug discovery. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
12. Going for broke: targeting the human cancer pseudokinome.
- Author
-
Bailey, Fiona P., Byrne, Dominic P., McSkimming, Daniel, Kannan, Natarajan, and Eyers, Patrick A.
- Subjects
- *
CELLULAR signal transduction , *SOMATIC mutation , *PHOSPHORYLATION , *PROTEIN kinases , *GENETIC code - Abstract
Protein phosphorylation lies at the heart of cell signalling, and somatic mutation(s) in kinases drives and sustains a multitude of human diseases, including cancer. The human protein kinase superfamily (the kinome) encodes approximately 50 'pseudokinases', which were initially predicted to be incapable of dynamic cell signalling when compared with canonical enzymatically active kinases. This assumption was supported by bioinformatics, which showed that amino acid changes at one or more key loci, making up the nucleotide-binding site or phosphotransferase machinery, were conserved in multiple vertebrate and non-vertebrate pseudokinase homologues. Protein kinases are highly attractive targets for drug discovery, as evidenced by the approval of almost 30 kinase inhibitors in oncology, and the successful development of the dual JAK1/2 (Janus kinase 1/2) inhibitor ruxolitinib for inflammatory indications. However, for such a large (>550) protein family, a remarkable number have still not been analysed at the molecular level, and only a surprisingly small percentage of kinases have been successfully targeted clinically. This is despite evidence that many are potential candidates for the development of new therapeutics. Indeed, several recent reports confirm that disease-associated pseudokinases can bind to nucleotide co-factors at concentrations achievable in the cell. Together, these findings suggest that drug targeting using either ATP-site or unbiased ligand-discovery approaches should now be attempted using the validation technology currently employed to evaluate their classic protein kinase counterparts. In the present review, we discuss members of the human pseudokinome repertoire, and catalogue somatic amino acid pseudokinase mutations that are emerging as the depth and clinical coverage of the human cancer pseudokinome expand. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
13. Day of the dead: pseudokinases and pseudophosphatases in physiology and disease.
- Author
-
Reiterer, Veronika, Eyers, Patrick A., and Farhan, Hesso
- Subjects
- *
KINASES , *PHOSPHATASES , *CELLULAR signal transduction , *LIGANDS (Biochemistry) , *TARGETED drug delivery - Abstract
Pseudophosphatases and pseudokinases are increasingly viewed as integral elements of signaling pathways, and there is mounting evidence that they have frequently retained the ability to interact with cellular ‘substrates’, and can exert important roles in different diseases. However, these pseudoenzymes have traditionally received scant attention compared to classical kinases and phosphatases. In this review we explore new findings in the emerging pseudokinase and pseudophosphatase fields, and discuss their different modes of action which include exciting new roles as scaffolds, anchors, spatial modulators, traps, and ligand-driven regulators of canonical kinases and phosphatases. Thus, it is now apparent that pseudokinases and pseudophosphatases both support and drive a panoply of signaling networks. Finally, we highlight recent evidence on their involvement in human pathologies, marking them as potential novel drug targets. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
14. The molecular regulation of Janus kinase (JAK) activation.
- Author
-
BABON, Jeffrey J., LUCET, Isabelle S., MURPHY, James M., NICOLA, Nicos A., and VARGHESE, Leila N.
- Subjects
- *
CYTOKINE receptors , *CELLULAR signal transduction , *PROTEIN-tyrosine kinases , *IMMUNODEFICIENCY , *INFLAMMATION , *GENETIC mutation - Abstract
The JAK (Janus kinase) family members serve essential roles as the intracellular signalling effectors of cytokine receptors. This family, comprising JAK1, JAK2, JAK3 and TYK2 (tyrosine kinase 2), was first described more than 20 years ago, but the complexities underlying their activation, regulation and pleiotropic signalling functions are still being explored. Here, we review the current knowledge of their physiological functions and the causative role of activating and inactivating JAK mutations in human diseases, including haemopoietic malignancies, immunodeficiency and inflammatory diseases. At the molecular level, recent studies have greatly advanced our knowledge of the structures and organization of the component FERM (4.1/ezrin/radixin/moesin)-SH2 (Src homology 2), pseudokinase and kinase domains within the JAKs, the mechanism of JAK activation and, in particular, the role of the pseudokinase domain as a suppressor of the adjacent tyrosine kinase domain's catalytic activity. We also review recent advances in our understanding of the mechanisms of negative regulation exerted by the SH2 domain-containing proteins, SOCS (suppressors of cytokine signalling) proteins and LNK. These recent studies highlight the diversity of regulatory mechanisms utilized by the JAK family to maintain signalling fidelity, and suggest alternative therapeutic strategies to complement existing ATP-competitive kinase inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
15. Pseudokinase drug intervention: a potentially poisoned chalice.
- Author
-
Claus, Jeroen, Cameron, Angus J. M., and Parker, Peter J.
- Subjects
- *
PROTEIN kinases , *ADENOSINE triphosphate , *PROTEIN-protein interactions , *EPIDERMAL growth factor receptors , *EUKARYOTIC cells , *KINASE inhibitors , *CANCER research - Abstract
Pseudokinases, the catalytically impaired component of the kinome, have recently been found to share more properties with active kinases than previously thought. In many pseudokinases, ATP binding and even some activity is preserved, highlighting these proteins as potential drug targets. In both active kinases and pseudokinases, binding of ATP or drugs in the nucleotide-binding pocket can stabilize specific conformations required for activity and protein-protein interactions. We discuss the implications of locking particular conformations in a selection of (pseudo)kinases and the dual potential impact on the druggability of these proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
16. The dual function of KSR1: a pseudokinase and beyond.
- Author
-
Hua Zhang, Chuay Yeng Koo, Stebbing, Justin, and Giamas, Georgios
- Subjects
- *
PROTEIN kinases , *CELL death , *CELLULAR signal transduction , *PHOSPHORYLATION , *DROSOPHILA , *CAENORHABDITIS elegans - Abstract
Protein kinases play a pivotal role in regulating many aspects of biological processes, including development, differentiation and cell death. Within the kinome, 48 kinases (∼10%) are classified as pseudokinases owing to the fact that they lack at least one conserved catalytic residue in their kinase domain. However, emerging evidence suggest that some pseudokinases, even without the ability to phosphorylate substrates, are regulators of multiple cellular signalling pathways. Among these is KSR1 (kinase suppressor of Ras 1), which was initially identified as a novel kinase in the Ras/Raf pathway. Subsequent studies showed that KSR1 mainly functions as a platform to assemble different cellular components thereby facilitating signal transduction. In the present article, we discuss recent findings regarding KSR1, indicating that it has dual activity as an active kinase as well as a pseudokinase/scaffolding protein. Moreover, the biological functions of KSR1 in human disorders, notably in malignancies, are also reviewed. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
17. New insights into the evolutionary conservation of the sole PIKK pseudokinase Tra1/TRRAP
- Author
-
Alberto Elías-Villalobos, Philippe Fort, Dominique Helmlinger, Centre de recherche en Biologie Cellulaire (CRBM), and Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 1 (UM1)
- Subjects
Protein Folding ,Transcription, Genetic ,Protein subunit ,[SDV]Life Sciences [q-bio] ,pseudokinase ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Biochemistry ,Conserved sequence ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,chaperones ,Amino Acid Sequence ,Transcription factor ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,0303 health sciences ,Sequence Homology, Amino Acid ,Kinase ,Nuclear Proteins ,Promoter ,Biological Evolution ,Cell biology ,Chromatin ,Phosphorylation ,chromatin ,Signal transduction ,transcription ,macromolecular complex 13 14 ,030217 neurology & neurosurgery ,Protein Binding ,Signal Transduction - Abstract
International audience; Phosphorylation by protein kinases is a fundamental mechanism of signal transduction. Many kinase families contain one or several members that, although evolutionarily conserved , lack the residues required for catalytic activity. Studies combining structural, biochemical , and functional approaches revealed that these pseudokinases have crucial roles in vivo and may even represent attractive targets for pharmacological intervention. Pseudokinases mediate signal transduction by a diversity of mechanisms, including allo-steric regulation of their active counterparts, assembly of signaling hubs, or modulation of protein localization. One such pseudokinase, named Tra1 in yeast and transformation/ transcription domain-associated protein (TRRAP) in mammals, is the only member lacking all catalytic residues within the phosphatidylinositol 3-kinase related kinase (PIKK) family of kinases. PIKKs are related to the PI3K family of lipid kinases, but function as Serine/Threonine protein kinases and have pivotal roles in diverse processes such as DNA damage sensing and repair, metabolic control of cell growth, nonsense-mediated decay, or transcription initiation. Tra1/TRRAP is the largest subunit of two distinct tran-scriptional co-activator complexes, SAGA and NuA4/TIP60, which it recruits to promoters upon transcription factor binding. Here, we review our current knowledge on the Tra1/ TRRAP pseudokinase, focusing on its role as a scaffold for SAGA and NuA4/TIP60 complex assembly and recruitment to chromatin. We further discuss its evolutionary history within the PIKK family and highlight recent findings that reveal the importance of molecular chaperones in pseudokinase folding, function, and conservation.
- Published
- 2019
- Full Text
- View/download PDF
18. Crystal structure of DRIK1, a stress-responsive receptor-like pseudokinase, reveals the molecular basis for the absence of ATP binding
- Author
-
Viviane C. H. da Silva, Katlin B. Massirer, Bruno Aquino, and Paulo Arruda
- Subjects
0106 biological sciences ,0301 basic medicine ,Drought stress ,Plant Science ,Biology ,01 natural sciences ,Zea mays ,Protein kinase ,03 medical and health sciences ,Adenosine Triphosphate ,Biotic stress ,Gene Expression Regulation, Plant ,Stress, Physiological ,lcsh:Botany ,Protein kinase A ,Gene ,Protein Kinase Inhibitors ,Plant Proteins ,Crystallography ,Abiotic stress ,Kinase ,Gene Expression Profiling ,Pseudokinase ,Receptor Protein-Tyrosine Kinases ,Small molecule ,Cell biology ,lcsh:QK1-989 ,030104 developmental biology ,Pyrimidines ,Protein kinase domain ,Phosphorylation ,Pyrazoles ,010606 plant biology & botany ,Research Article - Abstract
Background Plants reprogram metabolism and development to rapidly adapt to biotic and abiotic stress. Protein kinases play a significant role in this process by phosphorylating protein substrates that activate or inactivate signaling cascades that regulate cellular and metabolic adaptations. Despite their importance in plant biology, a notably small fraction of the plant kinomes has been studied to date. Results In this report, we describe ZmDRIK1, a stress-responsive receptor-like pseudokinase whose expression is downregulated under water restriction. We show the structural features and molecular basis of the absence of ATP binding exhibited by ZmDRIK1. The ZmDRIK1 kinase domain lacks conserved amino acids that are essential for phosphorylation activity. The crystal structure of the ZmDRIK1 kinase domain revealed the presence of a spine formed by the side chain of the triad Leu240, Tyr363, and Leu375 that occludes the ATP binding pocket. Although ZmDRIK1 is unable to bind nucleotides, it does bind the small molecule ENMD-2076 which, in a cocrystal structure, revealed the potential to serve as a ZmDRIK1 inhibitor. Conclusion ZmDRIK1 is a novel receptor-like pseudokinase responsive to biotic and abiotic stress. The absence of ATP binding and consequently, the absence of phosphorylation activity, was proven by the crystal structure of the apo form of the protein kinase domain. The expression profiling of the gene encoding ZmDRIK1 suggests this kinase may play a role in downregulating the expression of stress responsive genes that are not necessary under normal conditions. Under biotic and abiotic stress, ZmDRIK1 is down-regulated to release the expression of these stress-responsive genes.
- Published
- 2019
- Full Text
- View/download PDF
19. Correction for Beraki et al., Divergent kinase regulates membrane ultrastructure of the
- Subjects
PNAS Plus ,kinase ,phosphorylation ,pseudokinase ,chaperone ,Biological Sciences ,host–pathogen interaction ,Microbiology - Abstract
Significance Proteins are chemically modified after translation to regulate their functions. Protein kinases are enzymes that modify proteins with phosphate molecules. Using bioinformatics, we identified an unusual family of kinases (WNG kinases) that lack a structural motif, called the Gly-loop, which is absolutely required for the activity of all previously described kinases. Nevertheless, we found that the most conserved WNG kinase, WNG1, is catalytically active. The WNG kinases are only found in certain intracellular parasites, such as the human pathogen Toxoplasma gondii. We show that the founding member of the family, WNG1, phosphorylates parasite proteins that regulate membrane structure, and is therefore required for the proper biogenesis of the Toxoplasma parasitophorous vacuole, a structure essential for the parasite to cause disease., Apicomplexan parasites replicate within a protective organelle, called the parasitophorous vacuole (PV). The Toxoplasma gondii PV is filled with a network of tubulated membranes, which are thought to facilitate trafficking of effectors and nutrients. Despite being critical to parasite virulence, there is scant mechanistic understanding of the network’s functions. Here, we identify the parasite-secreted kinase WNG1 (With-No-Gly-loop) as a critical regulator of tubular membrane biogenesis. WNG1 family members adopt an atypical protein kinase fold lacking the glycine rich ATP-binding loop that is required for catalysis in canonical kinases. Unexpectedly, we find that WNG1 is an active protein kinase that localizes to the PV lumen and phosphorylates PV-resident proteins, several of which are essential for the formation of a functional intravacuolar network. Moreover, we show that WNG1-dependent phosphorylation of these proteins is required for their membrane association, and thus their ability to tubulate membranes. Consequently, WNG1 knockout parasites have an aberrant PV membrane ultrastructure. Collectively, our results describe a unique family of Toxoplasma kinases and implicate phosphorylation of secreted proteins as a mechanism of regulating PV development during parasite infection.
- Published
- 2019
20. Tribbles Pseudokinase 3 Regulation and Contribution to Cancer
- Author
-
Olivia Fromigué, Bojana Stefanovska, and Fabrice Andre
- Subjects
signaling pathway ,0301 basic medicine ,Scaffold protein ,Cancer Research ,Spliceosome ,RNA splicing ,tumor suppressor ,pseudokinase ,Review ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,oncogene ,Kinase activity ,Transcription factor ,biology ,rapamycin ,Kinase ,Akt ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,FK506 binding protein ,Cell biology ,030104 developmental biology ,Oncology ,TRIB3 ,030220 oncology & carcinogenesis ,mTOR ,biology.protein ,Signal transduction ,ER stress - Abstract
Simple Summary Accumulating evidence supports a key function for Tribbles proteins in oncogenesis, both in leukemia and solid tumors. However, the exact role of these proteins is hard to define since in a context-dependent manner they can function as both oncogenes and tumor suppressors. Their complex role arises from the capacity to interact with a wide range of target molecules thereby acting as molecular scaffolds and signaling regulators of multiple pathways. This review focuses on one particular Tribbles family member, namely, TRIB3, addressing its gene and protein expression, as well as its role in cancer development and progression. Abstract The first Tribbles protein was identified as critical for the coordination of morphogenesis in Drosophila melanogaster. Three mammalian homologs were subsequently identified, with a structure similar to classic serine/threonine kinases, but lacking crucial amino acids for the catalytic activity. Thereby, the very weak ATP affinity classifies TRIB proteins as pseudokinases. In this review, we provide an overview of the regulation of TRIB3 gene expression at both transcriptional and post-translational levels. Despite the absence of kinase activity, TRIB3 interferes with a broad range of cellular processes through protein–protein interactions. In fact, TRIB3 acts as an adaptor/scaffold protein for many other proteins such as kinase-dependent proteins, transcription factors, ubiquitin ligases, or even components of the spliceosome machinery. We then state the contribution of TRIB3 to cancer development, progression, and metastasis. TRIB3 dysregulation can be associated with good or bad prognosis. Indeed, as TRIB3 interacts with and regulates the activity of many key signaling components, it can act as a tumor-suppressor or oncogene in a context-dependent manner.
- Published
- 2021
- Full Text
- View/download PDF
21. Shared and distinct functions of the pseudokinase CORYNE (CRN) in shoot and root stem cell maintenance of Arabidopsis
- Author
-
Andrea Bleckmann, Rüdiger Simon, and Marc Somssich
- Subjects
0301 basic medicine ,Arabidopsis thaliana ,Physiology ,Arabidopsis ,pseudokinase ,Receptors, Cell Surface ,Plant Science ,Protein Serine-Threonine Kinases ,peptide signaling ,Plant Roots ,03 medical and health sciences ,Transduction (genetics) ,CORYNE ,CLAVATA ,Receptor ,biology ,stem cell maintenance ,Kinase ,Stem Cells ,fungi ,food and beverages ,Meristem maintenance ,Meristem ,biology.organism_classification ,Cell biology ,030104 developmental biology ,Protein kinase domain ,Signal transduction ,Plant Shoots ,Research Paper - Abstract
Highlight This study shows that the pseudokinase domain of the CLV2 co-receptor CRN is actively involved in transmitting the CLV3 peptide signal in the Arabidopsis shoot, but not in the proximal root meristem., Stem cell maintenance in plants depends on the activity of small secreted signaling peptides of the CLAVATA3/EMBRYO SURROUNDING REGION (CLE) family, which, in the shoot, act through at least three kinds of receptor complexes, CLAVATA1 (CLV1) homomers, CLAVATA2 (CLV2) / CORYNE (CRN) heteromers, and CLV1/CLV2/CRN multimers. In the root, the CLV2/CRN receptor complexes function in the proximal meristem to transmit signals from the CLE peptide CLE40. While CLV1 consists of an extracellular receptor domain and an intracellular kinase domain, CLV2, a leucine-rich repeat (LRR) receptor-like protein, and CRN, a protein kinase, have to interact to form a receptor–kinase complex. The kinase domain of CRN has been reported to be catalytically inactive, and it is not yet known how the CLV2/CRN complex can relay the perceived signal into the cells, and whether the kinase domain is necessary for signal transduction at all. In this study we show that the kinase domain of CRN is actively involved in CLV3 signal transduction in the shoot apical meristem of Arabidopsis, but it is dispensable for CRN protein function in root meristem maintenance. Hence, we provide an example of a catalytically inactive pseudokinase that is involved in two homologous pathways, but functions in distinctively different ways in each of them.
- Published
- 2016
22. Inhibitor-induced HER2-HER3 heterodimerisation promotes proliferation through a novel dimer interface
- Author
-
Paul R. Barber, Yosef Yarden, Roger George, Gargi Patel, Flavia Autore, Boris Vojnovic, Gregory Weitsman, Audrey Colomba, Marisa L. Martin-Fernandez, Selene K. Roberts, Peter J. Parker, Laura C. Zanetti-Domingues, Tony Ng, Michael Hirsch, Francesca Collu, Angus J.M. Cameron, Franca Fraternali, Tanya N. Soliman, Elena Ortiz-Zapater, and Jeroen Claus
- Subjects
0301 basic medicine ,Receptor, ErbB-3 ,Protein Conformation ,Receptor, ErbB-2 ,Adenosine Triphosphate ,ErbB2 ,Tumor Cells, Cultured ,kinase inhibitors ,Medicine ,FRET-FLIM ,ERBB3 ,Biology (General) ,Phosphorylation ,skin and connective tissue diseases ,Receptor ,Cancer Biology ,Kinase ,General Neuroscience ,protein kinase ,General Medicine ,3. Good health ,Female ,Research Article ,Human ,Signal Transduction ,medicine.drug ,QH301-705.5 ,Science ,Neuregulin-1 ,pseudokinase ,Breast Neoplasms ,Lapatinib ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Breast cancer ,HER3 ,HER2 ,Humans ,Protein kinase A ,Protein Kinase Inhibitors ,neoplasms ,Cell Proliferation ,General Immunology and Microbiology ,business.industry ,Cancer ,Cell Biology ,medicine.disease ,030104 developmental biology ,Cancer cell ,Cancer research ,Protein Multimerization ,business - Abstract
While targeted therapy against HER2 is an effective first-line treatment in HER2+ breast cancer, acquired resistance remains a clinical challenge. The pseudokinase HER3, heterodimerisation partner of HER2, is widely implicated in the resistance to HER2-mediated therapy. Here, we show that lapatinib, an ATP-competitive inhibitor of HER2, is able to induce proliferation cooperatively with the HER3 ligand neuregulin. This counterintuitive synergy between inhibitor and growth factor depends on their ability to promote atypical HER2-HER3 heterodimerisation. By stabilising a particular HER2 conformer, lapatinib drives HER2-HER3 kinase domain heterocomplex formation. This dimer exists in a head-to-head orientation distinct from the canonical asymmetric active dimer. The associated clustering observed for these dimers predisposes to neuregulin responses, affording a proliferative outcome. Our findings provide mechanistic insights into the liabilities involved in targeting kinases with ATP-competitive inhibitors and highlight the complex role of protein conformation in acquired resistance., eLife digest Around 20% of breast cancers are caused because cells have too many copies of a receptor protein called HER2 on their surface. HER2 is responsible for telling the cell to divide. Cells with too many of these receptors – and breast cancer cells can have up to 1000 times too many – divide uncontrollably. This causes the cancer to grow. Several successful anti-cancer drugs, such as Herceptin and Kadcyla, are used in the clinic to block the signals produced by HER2. Other drugs called kinase inhibitors prevent HER2 from building its faulty signals. However, a particular kinase inhibitor called lapatinib was not as successful in clinical trials as the medical community had hoped. Kinase inhibitors can have unexpected effects. While they can block specific signals in a cell, they can sometimes also cause new types of signals. Could this be one of the reasons behind the disappointing clinical trial results for lapatinib? By performing experiments on breast cancer cells grown in the laboratory, Claus, Patel et al. found that lapatinib can counterintuitively boost the growth of breast cancer cells. This occurs because lapatinib causes HER2 receptors to cluster together like a daisy chain along with another protein receptor of the same family, called HER3. These chains are primed to rapidly respond to a molecule called neuregulin, a growth factor that is commonly associated with breast cancer. The results presented by Claus, Patel et al. indicate that a particular subset of breast cancer patients – those whose cancer cells do not increase production of HER3 receptors – might better respond to lapatinib than others. The insights gained into what happens to HER2 when you try to block it should also influence the design of new drugs that target either HER2 or HER3.
- Published
- 2018
- Full Text
- View/download PDF
23. Hyperactivation of Oncogenic JAK3 Mutants Depend on ATP Binding to the Pseudokinase Domain
- Author
-
Juuli Raivola, Henrik M. Hammarén, Anniina T. Virtanen, Vilasha Bulleeraz, Alister C. Ward, Olli Silvennoinen, Lääketieteen ja biotieteiden tiedekunta - Faculty of Medicine and Life Sciences, University of Tampere, Institute of Biotechnology, and University of Helsinki
- Subjects
0301 basic medicine ,Cancer Research ,Cell signaling ,Biolääketieteet - Biomedicine ,3122 Cancers ,pseudokinase ,TYROSINE KINASE ,lcsh:RC254-282 ,03 medical and health sciences ,JANUS KINASES ,cytokine ,CRYSTAL-STRUCTURES ,Protein kinase A ,SPECIFICITY ,ACUTE LYMPHOBLASTIC-LEUKEMIA ,Original Research ,Common gamma chain ,ACTIVATING MUTATIONS ,COMPLEX ,Chemistry ,Kinase ,Janus kinase 3 ,leukemia ,PROTEIN-KINASE ,biochemical phenomena, metabolism, and nutrition ,TYK2 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,030104 developmental biology ,Oncology ,Phosphorylation ,nucleotide binding ,INHIBITORS ,Janus kinase ,Tyrosine kinase ,JAK kinase - Abstract
Janus kinase 3 (JAK3) tyrosine kinase has a central role in the control of lymphopoiesis, and mutations in JAK3 can lead to either severe combined immunodeficiency or leukemia and lymphomas. JAK3 associates with the common gamma chain (yc) receptor and functions in a heteromeric signaling pair with JAK1. In IL-2 signaling JAK1 is the effector kinase for STAT5 phosphorylation but the precise molecular regulatory mechanisms of JAK1 and JAK3 and their individual domains are not known. The pseudokinase domain (JAK homology 2, JH2) of JAK3 is of particular interest as approximately half of clinical JAK3 mutations cluster into it. In this study, we investigated the role of JH2s of JAK1 and JAK3 in IL-2R signaling and show that STAT5 activation requires both JH1 and JH2 of JAK1, while both JH1 and JH2 in JAK3 are specifically required for the cytokine-induction of cellular signaling. Characterization of recombinant JAK3 JH2 in thermal shift assay shows an unstable protein domain, which is strongly stabilized by ATP binding. Unexpectedly, nucleotide binding to JAK3 JH2 was found to be cation-independent. JAK3 JH2 showed higher nucleotide binding affinity in MANT-ATP and fluorescent polarization competition assays compared to the other JAK JH2s. Analysis of the functional role of ATP binding in JAK3 JH2 in cells and in zebrafish showed that disruption of ATP binding suppresses ligand-independent activation of clinical JAK3 gain-of-function mutations residing in either JH2 or JH1 but does not inhibit constitutive activation of oncogenic JAK1. ATP-binding site mutations in JAK3 JH2 do not, however, abrogate normal IL-2 signaling making them distinct from JH2 deletion or kinase-deficient JAK3. These findings underline the importance of JAK3 JH2 for cellular signaling in both ligand-dependent and in gain-of-function mutation-induced activation. Furthermore, they identify the JH2 ATP-binding site as a key regulatory region for oncogenic JAK3 signaling, and thus a potential target for therapeutic modulation.
- Published
- 2018
24. The SERK3 elongated allele defines a role for BIR ectodomains in brassinosteroid signalling
- Author
-
Andrea Moretti, Michael Hothorn, Ludwig A. Hothorn, Joël Nicolet, and Ulrich Hohmann
- Subjects
0106 biological sciences ,0301 basic medicine ,Protein Conformation ,Arabidopsis ,Leucine-rich repeat domain ,Plant Science ,Plasma protein binding ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,01 natural sciences ,Protein kinase ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Protein structure ,Brassinosteroids ,medicine ,Membrane receptor kinase ,Brassinosteroid ,Protein Interaction Domains and Motifs ,Receptor activation ,Receptor ,Alleles ,Plant Proteins ,Protein inhibitor ,Mutation ,Arabidopsis Proteins ,Kinase ,fungi ,Pseudokinase ,Membrane Proteins ,Brassinosteroid signaling ,Cell biology ,030104 developmental biology ,ddc:580 ,chemistry ,Ectodomain ,Signal transduction ,Protein Kinases ,Signal Transduction ,010606 plant biology & botany - Abstract
The leucine-rich repeat receptor kinase (LRR-RK) BRASSINOSTEROID INSENSITIVE 1 (BRI1) requires a shape-complementary SOMATIC EMBRYOGENESIS RECEPTOR KINASE (SERK) co-receptor for brassinosteroid sensing and receptor activation1. Interface mutations that weaken the interaction between receptor and co-receptor in vitro reduce brassinosteroid signalling responses2. The SERK3 elongated (elg) allele3-5 maps to the complex interface and shows enhanced brassinosteroid signalling, but surprisingly no tighter binding to the BRI1 ectodomain in vitro. Here, we report that rather than promoting the interaction with BRI1, the elg mutation disrupts the ability of the co-receptor to interact with the ectodomains of BRI1-ASSOCIATED-KINASE1 INTERACTING KINASE (BIR) receptor pseudokinases, negative regulators of LRR-RK signalling6. A conserved lateral surface patch in BIR LRR domains is required for targeting SERK co-receptors and the elg allele maps to the core of the complex interface in a 1.25 A BIR3-SERK1 structure. Collectively, our structural, quantitative biochemical and genetic analyses suggest that brassinosteroid signalling complex formation is negatively regulated by BIR receptor ectodomains.
- Published
- 2018
25. Janus-kinaasien molekyylinsisäinen säätely
- Author
-
Hammarén, Henrik, Lääketieteen ja biotieteiden tiedekunta - Faculty of Medicine and Life Sciences, and University of Tampere
- Subjects
myeloproliferatiivinen neoplasia ,kinase ,pseudokinaasi ,myeloproliferative neoplasm ,Lääketieteellinen biokemia - Medical Biochemistry ,cytokine ,kinaasi ,pseudokinase ,sytokiini ,JAK - Abstract
Janus-kinaasit (JAKit) ovat ei-reseptorisia tyrosiinikinaaseja, jotka välittävät yli 60 sytokiinin viestejä soluissa. Nämä viestit säätelevät lukuisia biologisia tapahtumia, kuten immuunijärjestelmän toimintaa, hematopoieesia, aineenvaihduntaa sekä kehitystä. JAKit ovat monidomeenisia proteeineja, joissa viestivää, aktiivista tyrosiinikinaasidomeenia (JH1) edeltää nk. pseudokinaasidomeeni (JH2). JH2 on ensisijaisen tärkeä JAK-aktiivisuuden säätelyssä. Domeenista onkin löydetty lukuisia kliinisesti merkittäviä mutaatioita, kuten autosomaalisia JAK2-mutaatioita, jotka aiheuttavat ligandiriippumatonta JAK-aktivaatiota ja siten johtavat hematopoieettisiin maligniteetteihin. Tässä työssä esitellyissä tutkimuksissa selvitettiin JH2:n sekä sen mutaatioiden toimintaa JAK-säätelyssä. Tutkimuksissa havaittiin, että kaikissa JAK JH2:ssa on toiminnallinen nukleotidinsitomistasku, joka kykenee sitomaan ATP:ta ja pienmolekyylisiä inhibiittoreita. Tärkeimpänä löydöksenä havaittiin, että ATP:n sitoutumisen estäminen JAK2 JH2:n ATP-sitomistaskuun alentaa ligandiriippumatonta JAK2-aktivaatiota. Nämä havainnot osoittavat, että JAK2 JH2 on mahdollinen lääkekohde kohdennettujen JAK-inhibiittoreiden kehittämiselle. Tällaiset inhibiittorit, jotka estäisivät kohdennetusti mutatoituneen (muttei villityyppisen) JAKin toimintaa, olisivat merkittävä parannus verrattuna nykyisiin JAK-estäjiin, jotka eivät kykene erottelemaan villityyppisen ja mutatoituneen JAKin välillä, eivätkä siten pysty parantamaan tautia. Tutkimuksissa kehitettiin yhteistyöprojektina myös molekyylimalli JH2:n toiminnasta JH1:n aktiivisuuden säätelijänä. JH2–JH1-malli selittää useimpien tunnettujen kliinisten JAK2-mutaatioiden toiminnan molekyylitasolla. Lisäksi suoritettiin systemaattinen analyysi JAK2-mutaatioista, jotka voivat estää tautia aiheuttavan JAK2-hyperaktivaation. Tämä analyysi tarkentaa ymmärrystämme JAKien toiminnasta sytokiinivälitteisessä sekä sytokiineista riippumattomassa JAK-aktivaatiossa ja samalla tunnistaa aiemmin tuntemattoman JAK2 JH2 rajapinnan, joka on välttämätön interferoni-γ signaloinnille. Janus kinases (JAKs) are non-receptor tyrosine kinases that mediate signalling of around sixty different cytokines governing various biological processes from the regulation of the immune system, to control of haematopoiesis, metabolism, and development. JAKs are multidomain proteins, in which the tyrosine kinase domain (JH1) is preceded by a pseudokinase domain (JH2). JH2 has critical regulatory functions and is a hotspot for many known oncogenic driver mutations. These mutations, which cause ligand-independent JAK activation, underlie various diseases—most notably haematopoietic malignancies caused by somatic JAK2 JH2 mutations. In the work presented here, we analysed the functions of JH2 and its mutations in the regulation of JAK activity. We found that all JAK JH2s have functional nucleotide-binding sites accessible to ATP and small molecule inhibitors. Most importantly, we found that disruption of ATP binding to JAK2 JH2 suppresses ligand-independent activation, thus identifying the JAK2 JH2 ATP-binding site as a potential drug target for the development of mutation-specific inhibitors. These inhibitors would be a distinct improvement over inhibitors of JAK2 currently used to treat MPNs, as current inhibitors do not distinguish between mutated and wild-type JAK2, and are unable to eradicate the disease. We also present a collaboration effort leading to a simulation-based model for JH2-mediated inhibition of JH1, thereby providing rationale for most known clinical JAK2 mutations. Moreover, we refine our understanding of ligand-mediated and ligand-independent activation of JAKs by presenting a systematic analysis of JAK2 mutations capable of inhibiting ligand-independent hyperactivation. We further identify a novel interface in JAK2 JH2, which is needed for heteromeric JAK2 activation in interferon-γ signalling.
- Published
- 2017
26. Nucleotide-binding mechanisms in pseudokinases
- Author
-
Henrik Hammaren, Anniina Virtanen, Olli Silvennoinen, Lääketieteen yksikkö - School of Medicine, and University of Tampere
- Subjects
0301 basic medicine ,Amino Acid Motifs ,Biophysics ,pseudokinase ,Review Article ,kinase activity ,Computational biology ,Biology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine Triphosphate ,Lääketieteen bioteknologia - Medical biotechnology ,Animals ,Humans ,Kinome ,Nucleotide ,signalling ,Kinase activity ,Binding site ,Review Articles ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,Kinase ,Mechanism (biology) ,Phosphotransferases ,kinome ,Cell Biology ,Protein Structure, Tertiary ,ATP ,030104 developmental biology ,chemistry ,nucleotide binding ,Adenosine triphosphate ,Function (biology) - Abstract
Pseudokinases are classified by the lack of one or several of the highly conserved motifs involved in nucleotide (nt) binding or catalytic activity of protein kinases (PKs). Pseudokinases represent ∼10% of the human kinome and they are found in all evolutionary classes of kinases. It has become evident that pseudokinases, which were initially considered somewhat peculiar dead kinases, are important components in several signalling cascades. Furthermore, several pseudokinases have been linked to human diseases, particularly cancer, which is raising interest for therapeutic approaches towards these proteins. The ATP-binding pocket is a well-established drug target and elucidation of the mechanism and properties of nt binding in pseudokinases is of significant interest and importance. Recent studies have demonstrated that members of the pseudokinase family are very diverse in structure as well as in their ability and mechanism to bind nts or perform phosphoryl transfer reactions. This diversity also precludes prediction of pseudokinase function, or the importance of nt binding for said function, based on primary sequence alone. Currently available data indicate that ∼40% of pseudokinases are able to bind nts, whereas only few are able to catalyse occasional phosphoryl transfer. Pseudokinases employ diverse mechanisms to bind nts, which usually occurs at low, but physiological, affinity. ATP binding serves often a structural role but in most cases the functional roles are not precisely known. In the present review, we discuss the various mechanisms that pseudokinases employ for nt binding and how this often low-affinity binding can be accurately analysed.
- Published
- 2016
27. Regulation of Vaccinia Virus Replication: a Story of Viral Mimicry and a Novel Antagonistic Relationship Between Vaccinia Kinase and Pseudokinase
- Author
-
Olson, Annabel T
- Subjects
- vaccinia virus, kinase, pseudokinase, B1 kinase, B12 pseudokinase, poxvirus, Biology, Life Sciences
- Abstract
Poxviruses employ sophisticated signaling pathways that thwart cellular defense mechanisms and simultaneously ensure viral factors are modulated properly. Yet, our understanding of these complex signaling networks are incomplete. For example, the vaccinia B1 kinase plays a vital role in inactivating the cellular antiviral factor BAF, and is suggested to orchestrate other pathways. B1 is highly conserved among poxviruses and exhibits a remarkable degree of similarity to VRKs, a family of cellular kinases, suggesting that the viral enzyme has evolved to mimic VRK activity. Indeed, B1 and VRKs have been demonstrated to target a shared substrate, the DNA binding protein BAF, elucidating a signaling pathway important for mitosis and the antiviral response. Our research further characterized the role of B1 during vaccinia infection to gain novel insights into its regulation and integration with cellular signaling pathways. We began by constructing and characterizing the first B1 deletion virus (ΔB1). Then using this virus, we tested the hypothesis that cellular VRKs can complement B1 function, and discovered a VRK2 role in facilitating DNA replication in the absence of B1. Study of the VRK2 mechanism revealed that B1 and VRK2 mediate DNA replication via an additional pathway that is BAF independent. We also utilized the ΔB1 virus in an experimental evolution assay to perform an unbiased search for suppressor mutations and identify novel pathways involving B1. Interestingly, our characterization of the adapted viruses reveals that mutations correlating with a loss of function of the vaccinia B12 pseudokinase provide a striking fitness enhancement to this virus. Next, B12 characterization showed a nuclear localization, unique for poxvirus proteins, that is related to its repressive function. Our data indicate that B12 is not a global repressor, but inhibits vaccinia replication in the absence of the B1 kinase. The mechanism of B12 partially depends on suppression of BAF antiviral activity. However, the parallel B12 pathway to restrict virus replication is less clear. Together, our studies of B1 and B12 present novel evidence that a paralogous kinase-pseudokinase pair can exhibit a unique epistatic relationship in a virus, and orchestrate yet-to-be-discovered nuclear events during infection. Advisor: Matthew S. Wiebe
- Published
- 2019
28. Molecular Mechanism of CCAAT-Enhancer Binding Protein Recruitment by the TRIB1 Pseudokinase
- Author
-
Peter D. Mace, Isabelle S Lucet, Weiwen Dai, James M. Murphy, Sam A. Jamieson, and Yoshio Nakatani
- Subjects
Kinase ,Constitutive Photomorphogenesis Protein 1 ,Amino Acid Motifs ,Molecular Sequence Data ,Plasma protein binding ,Biology ,Protein Serine-Threonine Kinases ,DNA-binding protein ,Structural Biology ,CCAAT-Enhancer-Binding Protein-alpha ,Humans ,Amino Acid Sequence ,Binding site ,Transcription factor ,Molecular Biology ,Tribbles ,Binding Sites ,Ccaat-enhancer-binding proteins ,CCAAT-enhancer binding protein ,TRIB1 ,Pseudokinase ,COP1 ,Intracellular Signaling Peptides and Proteins ,C/EBP ,Molecular biology ,Cell biology ,Ubiquitin ligase ,A-site ,Enhancer Elements, Genetic ,Protein kinase domain ,DFG motif ,biology.protein ,Protein Binding - Abstract
SummaryCCAAT-enhancer binding proteins (C/EBPs) are transcription factors that play a central role in the differentiation of myeloid cells and adipocytes. Tribbles pseudokinases govern levels of C/EBPs by recruiting them to the COP1 ubiquitin ligase for ubiquitination. Here, we present the first crystal structure of a Tribbles protein, which reveals a catalytically inactive TRIB1 pseudokinase domain with a unique adaptation in the αC helix. A second crystal structure and biophysical studies of TRIB1 with its C-terminal extension, which includes the COP1-binding motif, show that the C-terminal extension is sequestered at a site formed by the modified TRIB1 αC helix. In addition, we have identified and characterized the TRIB1 substrate-recognition sequence within C/EBPα, which is evolutionarily conserved in C/EBP transcription factors. Binding studies indicate that C/EBPα recruitment is weaker in the presence of the C-terminal COP1-binding motif, but the magnitude of this effect suggests that the two bind distinct rather directly overlapping binding sites.
- Published
- 2015
29. What is the point of pseudokinases?
- Author
-
Andrey S. Shaw and Saravanan Raju
- Subjects
Gene Expression ,Biochemistry ,Dental Enamel Proteins ,Extracellular matrix ,Mice ,Casein Kinase I ,Ameloblasts ,Biology (General) ,Phosphorylation ,chemistry.chemical_classification ,Extracellular Matrix Proteins ,Kinase ,General Neuroscience ,General Medicine ,Founder Effect ,Recombinant Proteins ,Lepidoptera ,Medicine ,Female ,Insight ,Ameloblast ,Signal Transduction ,QH301-705.5 ,Science ,Molecular Sequence Data ,pseudokinase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mammary Glands, Animal ,P70S6 kinase ,extracellular protein phosphorylation ,Animals ,Humans ,human ,Amino Acid Sequence ,mouse ,Sequence Homology, Amino Acid ,General Immunology and Microbiology ,secretory pathway phosphorylation ,tooth enamel formation ,Mice, Inbred C57BL ,Enzyme ,Gene Expression Regulation ,chemistry ,kinase complex ,Protein Kinases ,Sequence Alignment - Abstract
Although numerous extracellular phosphoproteins have been identified, the protein kinases within the secretory pathway have only recently been discovered, and their regulation is virtually unexplored. Fam20C is the physiological Golgi casein kinase, which phosphorylates many secreted proteins and is critical for proper biomineralization. Fam20A, a Fam20C paralog, is essential for enamel formation, but the biochemical function of Fam20A is unknown. Here we show that Fam20A potentiates Fam20C kinase activity and promotes the phosphorylation of enamel matrix proteins in vitro and in cells. Mechanistically, Fam20A is a pseudokinase that forms a functional complex with Fam20C, and this complex enhances extracellular protein phosphorylation within the secretory pathway. Our findings shed light on the molecular mechanism by which Fam20C and Fam20A collaborate to control enamel formation, and provide the first insight into the regulation of secretory pathway phosphorylation.
- Published
- 2015
- Full Text
- View/download PDF
30. A secretory kinase complex regulates extracellular protein phosphorylation
- Author
-
Vincent S. Tagliabracci, Meghdad Rahdar, Junyu Xiao, Jixin Cui, Jack E. Dixon, and Jianzhong Wen
- Subjects
Gene Expression ,Sequence Homology ,Inbred C57BL ,Biochemistry ,Mice ,Ameloblasts ,Protein phosphorylation ,Biology (General) ,Phosphorylation ,chemistry.chemical_classification ,Extracellular Matrix Proteins ,Enamel paint ,Kinase ,Casein Kinase I ,General Neuroscience ,General Medicine ,Mammary Glands ,Recombinant Proteins ,Founder Effect ,Cell biology ,Lepidoptera ,Amino Acid ,visual_art ,visual_art.visual_art_medium ,Medicine ,Female ,Signal Transduction ,Research Article ,QH301-705.5 ,1.1 Normal biological development and functioning ,Science ,Molecular Sequence Data ,pseudokinase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,stomatognathic system ,Dental Enamel Proteins ,Underpinning research ,Extracellular ,biochemistry ,extracellular protein phosphorylation ,Animals ,Humans ,Amino Acid Sequence ,human ,Secretory pathway ,mouse ,General Immunology and Microbiology ,Animal ,secretory pathway phosphorylation ,tooth enamel formation ,Enzyme ,Secretory protein ,chemistry ,Gene Expression Regulation ,kinase complex ,Generic health relevance ,Biochemistry and Cell Biology ,Sequence Alignment - Abstract
Although numerous extracellular phosphoproteins have been identified, the protein kinases within the secretory pathway have only recently been discovered, and their regulation is virtually unexplored. Fam20C is the physiological Golgi casein kinase, which phosphorylates many secreted proteins and is critical for proper biomineralization. Fam20A, a Fam20C paralog, is essential for enamel formation, but the biochemical function of Fam20A is unknown. Here we show that Fam20A potentiates Fam20C kinase activity and promotes the phosphorylation of enamel matrix proteins in vitro and in cells. Mechanistically, Fam20A is a pseudokinase that forms a functional complex with Fam20C, and this complex enhances extracellular protein phosphorylation within the secretory pathway. Our findings shed light on the molecular mechanism by which Fam20C and Fam20A collaborate to control enamel formation, and provide the first insight into the regulation of secretory pathway phosphorylation. DOI: http://dx.doi.org/10.7554/eLife.06120.001, eLife digest Some proteins must be modified in order to work effectively. One common modification is to add a phosphate group to the protein, which is performed by enzymes called protein kinases. Although most of the protein kinases work on proteins inside the cell, it was discovered recently that a small group of kinases work within the ‘secretory pathway’ and modify proteins that are released (or secreted) out of cells. One such secretory pathway kinase—called Fam20C—phosphorylates a wide range of secreted proteins and helps to ensure the proper development of bones and teeth. Specifically, Fam20C and a closely related protein called Fam20A are important for forming enamel, the hardest substance in human body, which makes up the outer surface of teeth. However, the exact role of Fam20A is unknown. Cui et al. now show that Fam20A binds to Fam20C, and this increases the ability of Fam20C to phosphorylate the proteins that form the ‘matrix’ that guides the deposition of the enamel minerals. Furthermore, mutations in Fam20A lead to the inefficient phosphorylation of enamel matrix proteins by Fam20C, and prevent proper enamel formation. The results raise the possibility that similar mechanisms of secretory kinase activation may also be important in other biological processes where many secreted proteins need to be phosphorylated rapidly. DOI: http://dx.doi.org/10.7554/eLife.06120.002
- Published
- 2015
31. The many-faced KSR1: a tumor suppressor in breast cancer
- Author
-
Hua Zhang, Justin Stebbing, and Georgios Giamas
- Subjects
MAPK/ERK pathway ,Cancer Research ,Oncogene ,Kinase ,KSR1 ,pseudokinase ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Q1 ,Editorial ,breast cancer ,Breast cancer ,Oncology ,BARD1 ,medicine ,Cancer research ,Carcinogenesis - Abstract
Emerging evidence supports the dual function of kinase suppressor of Ras 1 (KSR1) as an active kinase and a scaffold, although it has been extensively referred as a pseudokinase, due to the absence of key residues in its catalytic domain [1, 2]. As a scaffolding protein, KSR1 orchestrates the assembly of the protein kinases RAF, mitogen activated protein kinase (MAPK) kinase (MEK), and extracellular signal-regulated kinase (ERK) in the canonical Ras-RAF-MAPKs pathway, in a Ras-dependent manner or upon growth factor treatment [1, 3]. Conversely, structural and biochemical studies reveal that KSR1 is capable of phosphorylating MEK and more importantly, the catalytic activity of KSR is markedly increased when BRAF or inhibitor-bound CRAF is introduced in the complexes [1, 4, 5]. Such findings add complexity to the ERK spatio-temporal pathway control and identify KSR1 as a modulator of these pathways. In light of its regulatory role in oncogenic Ras-RAF-MAPKs signaling, extensive efforts have attempted to establish KSR1 as an oncogene in Ras-dependent cancers. Indeed, KSR1 has been shown to contribute to oncogenesis in various forms of Ras-activated cancer, including skin, pancreatic and lung carcinomas [1]. First, absence of KSR1 inhibits tumor formation in different Ras-mediated mouse models (KSR1−/−), suggesting that KSR1 is required for Ras-transduced MAPK activated tumorigenesis. Moreover, depletion of KSR1 reduces tumor growth of K-Ras-dependent pancreatic and lung cancer xenografts in nude mice, further supporting KSR1 as an oncogene as well as a potential therapeutic target. However, our own studies reveal an interesting, and yet unexpected role of KSR1 in breast cancer, where Ras mutations are rare. Using tumor tissue microarrays in a large patient cohort with a long term follow-up, we observe that breast cancer patients with high KSR1 had better disease free- and overall survival, results also supported by Oncomine analyses, microarray data and genomic data from paired tumor and cell-free DNA (cfDNA) samples revealing loss of heterozygosity [6]. KSR1 expression is positively associated with breast cancer 1, early onset (BRCA1), BRCA1-associated ring domain 1 (BARD1) and checkpoint kinase 1 (Chk1) levels in breast cancer specimens. Intriguingly, phospho-profiling of major components of the canonical Ras-RAF-MAPKs pathway, including RAF, MEK and ERK, show no significant changes upon KSR1 overexpression or depletion. These results underline its role beyond coordinating MAPKs signaling and challenge its oncogenic function in breast cancer. Further experiments support a tumor suppressive role of KSR1. Of note, breast cancer cells overexpressing KSR1 form fewer and smaller size colonies compared to the parental ones, while an in vivo mouse model also demonstrates that the growth of xenograft tumors overexpressing KSR1 is inhibited. It appears that the tumor inhibitory effect of KSR1 is BRCA1-dependent as shown by in vitro 3D-matrigel and soft-agar assays. Consistent with the correlation between KSR1 and BRCA1 in clinical samples, in vitro assays demonstrate KSR1 overexpression increases BRCA1 protein levels by decreasing BRCA1 ubiquitination partly through elevating BARD1 protein abundance and the BRCA1-BARD1 interaction. These findings integrate KSR1 into a complicated signaling network involving BRCA1-BARD1 complex. Unquestionably, alternative mechanisms underlying BRCA1's up-regulation by KSR1 other than through BARD1 remain to be examined. Our current understanding of KSR1's function in breast cancer is far from complete (Figure (Figure1).1). Puzzlingly, in addition to executing its anti-tumor effects through BRCA1, KSR1 negatively regulates transcriptional activity of p53 by reducing p53 acetylation via modulation of deleted in breast cancer 1 (DBC1) phosphorylation [7]. How can KSR1 up-regulates BRCA1 and inhibits p53 activity at the same time? What determines its preferable binding partners and thus contributes to its inhibitory outcome? An interesting and simple theory might be the following: since DBC1 is a positive regulator of p53 activity and a repressor for BRCA1, it potentially positions KSR1 into a dynamic state connecting p53 and BRCA1. Ultimately, while BRCA1 compensates for the loss of p53 activity, KSR1 tilts the balance scale towards tumor suppression in cancer cells. Further investigation is much needed to address the underlying mechanisms. In addition, evidence regarding other aspects of its action are evolving: a very recent study characterizing the proteomic profile of KSR1-regulated signaling in response to genotoxic agents in breast cancer illustrates a broad functional network conferred by KSR1, highlighting its importance in the chemotherapy response [8].. Figure 1 KSR1-regulated signaling in cancer A better understanding of the molecular determinants of the distinctive behaviour of KSR1 in different types of cancer is essential to the optimal targeting of this dual functional kinase-scaffold protein. Our studies suggest the tumor suppressive action of KSR1 and its clinical relevance in patient stratification, placing KSR1 in the major oncoprotein pathways.
- Published
- 2015
32. HSP90 activity is required for MLKL oligomerisation and membrane translocation and the induction of necroptotic cell death
- Author
-
Joanne M Hildebrand, David C.S. Huang, Kym N Lowes, James M. Murphy, Zikou Liu, J-G Zhang, Isabelle S Lucet, Annette V. Jacobsen, Maria C. Tanzer, Stephanie A. Conos, John Silke, Guillaume Lessene, M F van Delft, Emma J. Petrie, Jacobsen, AV, Lowes, KN, Tanzer, MC, Lucet, IS, Hildebrand, Joan, Petrie, EJ, van Delft, MF, Liu, Z, Conos, SA, Zhang, J, Huang, DCS, Silke, J, Lessene, G, and Murphy, JM
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,heat-shock protein 90 ,Necroptosis ,Immunology ,necroptosis ,pseudokinase ,Apoptosis ,Translocation, Genetic ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,RIPK1 ,Mice ,Necrosis ,tumour necrosis factor ,Animals ,Humans ,Integrin-linked kinase ,HSP90 Heat-Shock Proteins ,Phosphorylation ,protein translocation ,biology ,Cell Death ,Effector ,Kinase ,cell signalling ,Cell Biology ,Hsp90 ,Cell biology ,030104 developmental biology ,biology.protein ,Cancer research ,Original Article ,Protein Kinases - Abstract
Necroptosis is a caspase-independent form of regulated cell death that has been implicated in the development of a range of inflammatory, autoimmune and neurodegenerative diseases. The pseudokinase, Mixed Lineage Kinase Domain-Like (MLKL), is the most terminal known obligatory effector in the necroptosis pathway, and is activated following phosphorylation by Receptor Interacting Protein Kinase-3 (RIPK3). Activated MLKL translocates to membranes, leading to membrane destabilisation and subsequent cell death. However, the molecular interactions governing the processes downstream of RIPK3 activation remain poorly defined. Using a phenotypic screen, we identified seven heat-shock protein 90 (HSP90) inhibitors that inhibited necroptosis in both wild-type fibroblasts and fibroblasts expressing an activated mutant of MLKL. We observed a modest reduction in MLKL protein levels in human and murine cells following HSP90 inhibition, which was only apparent after 15 h of treatment. The delayed reduction in MLKL protein abundance was unlikely to completely account for defective necroptosis, and, consistent with this, we also found inhibition of HSP90 blocked membrane translocation of activated MLKL. Together, these findings implicate HSP90 as a modulator of necroptosis at the level of MLKL, a function that complements HSP90’s previously demonstrated modulation of the upstream necroptosis effector kinases, RIPK1 and RIPK3.
- Published
- 2015
33. Titin kinase is an inactive pseudokinase scaffold that supports MuRF1 recruitment to the sarcomeric M-line
- Author
-
Daniel J. Rigden, Siegfried Labeit, Alexander Gasch, Olga Mayans, Julijus Bogomolovas, and Felix Simkovic
- Subjects
Models, Molecular ,Sarcomeres ,Myofilament ,Protein Conformation ,Immunology ,Muscle Proteins ,pseudokinase ,Spodoptera ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Evolution, Molecular ,phosphorylation assay ,Mice ,Protein structure ,Ubiquitin ,Catalytic Domain ,ddc:570 ,Sf9 Cells ,Animals ,Humans ,Connectin ,titin ,Amino Acid Sequence ,lcsh:QH301-705.5 ,Phylogeny ,biology ,Kinase ,General Neuroscience ,Research ,Ubiquitination ,Muscle, Striated ,Ubiquitin ligase ,Cell biology ,Protein Structure, Tertiary ,lcsh:Biology (General) ,Biochemistry ,Protein kinase domain ,biology.protein ,Titin ,Myofibril ,Sequence Alignment ,mutagenesis ,Research Article - Abstract
Striated muscle tissues undergo adaptive remodelling in response to mechanical load. This process involves the myofilament titin and, specifically, its kinase domain (TK; titin kinase) that translates mechanical signals into regulatory pathways of gene expression in the myofibril. TK mechanosensing appears mediated by a C-terminal regulatory tail (CRD) that sterically inhibits its active site. Allegedly, stretch-induced unfolding of this tail during muscle function releases TK inhibition and leads to its catalytic activation. However, the cellular pathway of TK is poorly understood and substrates proposed to date remain controversial. TK's best-established substrate is Tcap, a small structural protein of the Z-disc believed to link TK to myofibrillogenesis. Here, we show that TK is a pseudokinase with undetectable levels of catalysis and, therefore, that Tcap is not its substrate. Inactivity is the result of two atypical residues in TK's active site, M34 and E147, that do not appear compatible with canonical kinase patterns. While not mediating stretch-dependent phospho-transfers, TK binds the E3 ubiquitin ligase MuRF1 that promotes sarcomeric ubiquitination in a stress-induced manner. Given previous evidence of MuRF2 interaction, we propose that the cellular role of TK is to act as a conformationally regulated scaffold that functionally couples the ubiquitin ligases MuRF1 and MuRF2, thereby coordinating muscle-specific ubiquitination pathways and myofibril trophicity. Finally, we suggest that an evolutionary dichotomy of kinases/pseudokinases has occurred in TK-like kinases, where invertebrate members are active enzymes but vertebrate counterparts perform their signalling function as pseudokinase scaffolds.
- Published
- 2014
34. STRAD pseudokinases regulate axogenesis and LKB1 stability
- Author
-
Biliana O. Veleva-Rotse, Alexandria P. Gardner, Gabrielle Reilly, Krishnaveni Subbiah, Allyson Brown, Hans Clevers, Benjamin D. Edmonds, Eric A. Gaucher, Anthony P. Barnes, Zi ming Zhao, Annette F. Baas, Kelly Hansen, James L. Smart, Lillian R. Klug, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
LKB1 ,Neurodevelopment ,Morphogenesis ,Regulator ,Biology ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Axon ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Developmental Neuroscience ,Cell polarity ,medicine ,Animals ,030304 developmental biology ,Cerebral Cortex ,Neurons ,0303 health sciences ,Kinase ,Pseudokinase ,Adaptor Proteins ,Compartmentalization (psychology) ,Protein-Serine-Threonine Kinases ,STRAD ,Axons ,Cell biology ,Vesicular Transport ,Adaptor Proteins, Vesicular Transport ,medicine.anatomical_structure ,Cytoplasm ,Developmental biology ,Neuroscience ,030217 neurology & neurosurgery ,Research Article - Abstract
BACKGROUND: Neuronal polarization is an essential step of morphogenesis and connectivity in the developing brain. The serine/threonine kinase LKB1 is a key regulator of cell polarity, metabolism, tumorigenesis, and is required for axon formation. It is allosterically regulated by two related and evolutionarily conserved pseudokinases, STe20-Related ADapters (STRADs) α and β. The roles of STRADα and STRADβ in the developing nervous system are not fully defined, nor is it known whether they serve distinct functions. RESULTS: We find that STRADα is highly spliced and appears to be the primal STRAD paralog. We report that each STRAD is sufficient for axogenesis and promoting cell survival in the developing cortex. We also reveal a reciprocal protein-stabilizing relationship in vivo between LKB1 and STRADα, whereby STRADα specifically maintains LKB1 protein levels via cytoplasmic compartmentalization. CONCLUSIONS: We demonstrate a novel role for STRADβ in axogenesis and also show for the first time in vivo that STRADα, but not STRADβ, is responsible for LKB1 protein stability.
- Published
- 2013
35. A robust methodology to subclassify pseudokinases based on their nucleotide-binding properties
- Author
-
Jun Qin, Ling Liu, Isabelle S Lucet, Natalia Jura, Samuel N. Young, Patrick A. Eyers, Jeffrey J. Babon, Qingwei Zhang, Anne Tripaydonis, Leila N. Varghese, Daniela Ungureanu, Michael L. Reese, Zachary L. Nimchuk, Christine L. Gee, Koichi Fukuda, Roger J. Daly, Fiona P. Bailey, James M. Murphy, Henrik Hammaren, Gerard Manning, Mary Beth Mudgett, Kelan Chen, Sabine Elowe, and Olli Silvennoinen
- Subjects
Biochemistry & Molecular Biology ,Cation binding ,Insecta ,Receptor, ErbB-3 ,1.1 Normal biological development and functioning ,Molecular Sequence Data ,pseudokinase ,Plasma protein binding ,Biology ,Real-Time Polymerase Chain Reaction ,Medical and Health Sciences ,Biochemistry ,SH3 domain ,Article ,Cell Line ,Underpinning research ,ErbB-3 ,Animals ,Humans ,Nucleotide ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,non-catalytic protein-interaction domain ,Cancer ,chemistry.chemical_classification ,Kinase ,protein kinase ,Cell Biology ,Biological Sciences ,Janus Kinase 2 ,chemistry ,pseudoenzyme ,Chemical Sciences ,Cyclin-dependent kinase complex ,Generic health relevance ,nucleotide binding ,Function (biology) ,Receptor ,Protein Binding - Abstract
Protein kinase-like domains that lack conserved residues known to catalyse phosphoryl transfer, termed pseudokinases, have emerged as important signalling domains across all kingdoms of life. Although predicted to function principally as catalysis-independent protein-interaction modules, several pseudokinase domains have been attributed unexpected catalytic functions, often amid controversy. We established a thermal-shift assay as a benchmark technique to define the nucleotide-binding properties of kinase-like domains. Unlike in vitro kinase assays, this assay is insensitive to the presence of minor quantities of contaminating kinases that may otherwise lead to incorrect attribution of catalytic functions to pseudokinases. We demonstrated the utility of this method by classifying 31 diverse pseudokinase domains into four groups: devoid of detectable nucleotide or cation binding; cation-independent nucleotide binding; cation binding; and nucleotide binding enhanced by cations. Whereas nine pseudokinases bound ATP in a divalent cation-dependent manner, over half of those examined did not detectably bind nucleotides, illustrating that pseudokinase domains predominantly function as non-catalytic protein-interaction modules within signalling networks and that only a small subset is potentially catalytically active. We propose that henceforth the thermal-shift assay be adopted as the standard technique for establishing the nucleotide-binding and catalytic potential of kinase-like domains.
- Published
- 2013
36. Involvement of Lyn and the Atypical Kinase SgK269/PEAK1 in a Basal Breast Cancer Signaling Pathway
- Author
-
Brigid C. Browne, Howard Cheuk Ho Chan, David Gallego-Ortega, Danny Rickwood, Naveid A. Ali, Ling Liu, Luxi Zhang, Carole M Tactacan, Darren N. Saunders, Roger J. Daly, David R. Croucher, Falko Hochgräfe, Ruth J. Lyons, Alexander Swarbrick, and Robert F. Shearer
- Subjects
Cancer Research ,Breast Neoplasms ,Signal transduction ,Polymerase Chain Reaction ,Basal (phylogenetics) ,Breast cancer ,LYN ,Cell Line, Tumor ,medicine ,Grb2 ,Humans ,Neoplasm Invasiveness ,Phosphorylation ,skin and connective tissue diseases ,STAT3 ,biology ,Stat3 ,Kinase ,Pseudokinase ,tyrosine kinase ,Protein-Tyrosine Kinases ,medicine.disease ,src-Family Kinases ,Microscopy, Fluorescence ,Oncology ,biology.protein ,Cancer research ,Female ,GRB2 ,Triple negative ,Tyrosine kinase ,Cell Division ,Signal Transduction - Abstract
Basal breast cancer cells feature high expression of the Src family kinase Lyn that has been implicated in the pathogenicity of this disease. In this study, we identified novel Lyn kinase substrates, the most prominent of which was the atypical kinase SgK269 (PEAK1). In breast cancer cells, SgK269 expression associated with the basal phenotype. In primary breast tumors, SgK269 overexpression was detected in a subset of basal, HER2-positive, and luminal cancers. In immortalized MCF-10A mammary epithelial cells, SgK269 promoted transition to a mesenchymal phenotype and increased cell motility and invasion. Growth of MCF-10A acini in three-dimensional (3D) culture was enhanced upon SgK269 overexpression, which induced an abnormal, multilobular acinar morphology and promoted extracellular signal–regulated kinase (Erk) and Stat3 activation. SgK269 Y635F, mutated at a major Lyn phosphorylation site, did not enhance acinar size or cellular invasion. We show that Y635 represents a Grb2-binding site that promotes both Stat3 and Erk activation in 3D culture. RNA interference–mediated attenuation of SgK269 in basal breast cancer cells promoted acquisition of epithelial characteristics and decreased anchorage-independent growth. Together, our results define a novel signaling pathway in basal breast cancer involving Lyn and SgK269 that offers clinical opportunities for therapeutic intervention. National Health and Medical Research Council of Australia, Cancer Council New South Wales (NSW) and Science Foundation Ireland (Grant No. 06/CE/B1129). DRC and DS were supported by Fellowships from Cancer Institute (CI) NSW. FH is supported by The Ministry of Education and Research, Bundesministerium für Bildung und Forschung (03Z1CN21). CMT is the recipient of a Research Scholarship from CINSW, and LZ an Australian Postgraduate Award. DGO is supported by a National Breast Cancer Foundation (NBCF) and Cure Cancer Australia Foundation Postdoctoral Fellowship. AS is supported by an Early Career Fellowship from the NBCF. Deposited by bulk import
- Published
- 2013
37. Cell Fate Regulation Governed by a Repurposed Bacterial Histidine Kinase
- Author
-
Irimpan I. Mathews, W. Seth Childers, Lucy Shapiro, Thomas H. Mann, Jimmy A. Blair, Qingping Xu, and Ashley M. Deacon
- Subjects
Cell division ,Histidine Kinase ,Applied Microbiology ,Cell ,Applied Microbiology and Biotechnology ,0302 clinical medicine ,Cell Signaling ,Asymmetric cell division ,Biology (General) ,lcsh:QH301-705.5 ,Caulobacter Crescentus ,0303 health sciences ,Crystallography ,Protein Kinase Signaling Cascade ,biology ,Kinase ,Physics ,General Neuroscience ,Cell Cycle ,Cell cycle ,Condensed Matter Physics ,Signaling Cascades ,Bacterial Biochemistry ,Cell biology ,medicine.anatomical_structure ,Biochemistry ,two-component systems (TCS) ,Physical Sciences ,Crystal Structure ,Prokaryotic Models ,Phosphorylation ,Signal transduction ,Cellular Structures and Organelles ,General Agricultural and Biological Sciences ,Dimerization ,Protein Binding ,Research Article ,Signal Transduction ,QH301-705.5 ,Allosteric regulation ,pseudokinase ,response regulator ,Signaling Complexes ,Research and Analysis Methods ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Microbiology ,asymmetric cell division ,General Biochemistry, Genetics and Molecular Biology ,Caulobacter ,03 medical and health sciences ,Model Organisms ,Bacterial Proteins ,Virology ,medicine ,Genetics ,Solid State Physics ,Kinase activity ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Bacteria ,General Immunology and Microbiology ,Caulobacter crescentus ,Histidine kinase ,Organisms ,Biology and Life Sciences ,Bacteriology ,Cell Biology ,biology.organism_classification ,Protein Structure, Tertiary ,Response regulator ,lcsh:Biology (General) ,cell-cycle ,Cell Signaling Structures ,Parasitology ,Protein Kinases ,030217 neurology & neurosurgery - Abstract
The pathway that regulates asymmetric cell division in Caulobacter involves a signaling kinase whose catalytic output domain has been repurposed as an input sensor of the phosphorylation state of the response regulator – a reversal of the conventional direction of information flow; this allows wiring of simple linear signaling pathways into complex eukaryote-like networks., One of the simplest organisms to divide asymmetrically is the bacterium Caulobacter crescentus. The DivL pseudo-histidine kinase, positioned at one cell pole, regulates cell-fate by controlling the activation of the global transcription factor CtrA via an interaction with the response regulator (RR) DivK. DivL uniquely contains a tyrosine at the histidine phosphorylation site, and can achieve these regulatory functions in vivo without kinase activity. Determination of the DivL crystal structure and biochemical analysis of wild-type and site-specific DivL mutants revealed that the DivL PAS domains regulate binding specificity for DivK∼P over DivK, which is modulated by an allosteric intramolecular interaction between adjacent domains. We discovered that DivL's catalytic domains have been repurposed as a phosphospecific RR input sensor, thereby reversing the flow of information observed in conventional histidine kinase (HK)-RR systems and coupling a complex network of signaling proteins for cell-fate regulation., Author Summary Across all kingdoms of life the generation of cell-type diversity is the consequence of asymmetry at the point of cell division. The bacterium Caulobacter crescentus divides asymmetrically to produce daughter cells that have distinct morphology and behavior. As in eukaryotes, an unequal distribution of signaling proteins in daughter Caulobacter cells triggers the differential read-out of identical genomes. A critical interaction between two protein molecules – a protein kinase (DivL) and a response regulator (DivK) – is known to occur exclusively in one daughter cell and to thereby regulate differentiation. However, mapping the observed signaling interconnections that drive asymmetric division has been difficult to reconcile with traditional models of bacterial signaling. Here we determine how DivL detects and processes this DivK signal. Although DivL has an architecture that is typical of histidine kinases, which normally act by regulating the phosphorylation state of the appropriate response regulator, DivL's essential functions do not require kinase activity and DivL does not add or remove phosphate from DivK. Instead we find that DivL has converted its output kinase domain into an input sensor domain that specifically detects phosphorylated DivK, and we identify key features of DivL that underlie this specificity. This novel reassignment of sensory functions reverses the conventional kinase-to-response-regulator signaling flow and logically couples linear signaling pathways into complex eukaryote-like networks to regulate cell development.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.