Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell ; 179(1): 205-218.e21, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31522888

ABSTRACT

The molecular chaperone HSP90 facilitates the folding of several client proteins, including innate immune receptors and protein kinases. HSP90 is an essential component of plant and animal immunity, yet pathogenic strategies that directly target the chaperone have not been described. Here, we identify the HopBF1 family of bacterial effectors as eukaryotic-specific HSP90 protein kinases. HopBF1 adopts a minimal protein kinase fold that is recognized by HSP90 as a host client. As a result, HopBF1 phosphorylates HSP90 to completely inhibit the chaperone's ATPase activity. We demonstrate that phosphorylation of HSP90 prevents activation of immune receptors that trigger the hypersensitive response in plants. Consequently, HopBF1-dependent phosphorylation of HSP90 is sufficient to induce severe disease symptoms in plants infected with the bacterial pathogen, Pseudomonas syringae. Collectively, our results uncover a family of bacterial effector kinases with toxin-like properties and reveal a previously unrecognized betrayal mechanism by which bacterial pathogens modulate host immunity.


Subject(s)
Arabidopsis Proteins/metabolism , Bacterial Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Mimicry/immunology , Plant Immunity/physiology , Adenosine Triphosphatases/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/microbiology , Bacterial Proteins/chemistry , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HeLa Cells , Host Microbial Interactions/immunology , Humans , Phosphorylation , Plasmids/genetics , Protein Binding , Protein Folding , Protein Kinases/metabolism , Pseudomonas syringae/metabolism , Saccharomyces cerevisiae/metabolism
2.
Cell ; 175(3): 809-821.e19, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30270044

ABSTRACT

Approximately 10% of human protein kinases are believed to be inactive and named pseudokinases because they lack residues required for catalysis. Here, we show that the highly conserved pseudokinase selenoprotein-O (SelO) transfers AMP from ATP to Ser, Thr, and Tyr residues on protein substrates (AMPylation), uncovering a previously unrecognized activity for a member of the protein kinase superfamily. The crystal structure of a SelO homolog reveals a protein kinase-like fold with ATP flipped in the active site, thus providing a structural basis for catalysis. SelO pseudokinases localize to the mitochondria and AMPylate proteins involved in redox homeostasis. Consequently, SelO activity is necessary for the proper cellular response to oxidative stress. Our results suggest that AMPylation may be a more widespread post-translational modification than previously appreciated and that pseudokinases should be analyzed for alternative transferase activities.


Subject(s)
Adenosine Monophosphate/metabolism , Catalytic Domain , Protein Processing, Post-Translational , Selenoproteins/metabolism , Conserved Sequence , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Stress , Selenoproteins/chemistry
3.
Mol Cell ; 83(21): 3758-3760, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37922868

ABSTRACT

The SARS-CoV-2 NiRAN domain is essential for viral replication. Despite adopting a pseudokinase fold, it catalyzes three distinct biochemical reactions from a single active site. In this issue of Molecular Cell, Small et al.1 elucidate the structural intricacies of the NiRAN domain shedding light on the factors that underlie its remarkable versatility.


Subject(s)
SARS-CoV-2 , Virus Replication , Catalytic Domain
4.
Cell ; 161(7): 1619-32, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091039

ABSTRACT

The existence of extracellular phosphoproteins has been acknowledged for over a century. However, research in this area has been undeveloped largely because the kinases that phosphorylate secreted proteins have escaped identification. Fam20C is a kinase that phosphorylates S-x-E/pS motifs on proteins in milk and in the extracellular matrix of bones and teeth. Here, we show that Fam20C generates the majority of the extracellular phosphoproteome. Using CRISPR/Cas9 genome editing, mass spectrometry, and biochemistry, we identify more than 100 secreted phosphoproteins as genuine Fam20C substrates. Further, we show that Fam20C exhibits broader substrate specificity than previously appreciated. Functional annotations of Fam20C substrates suggest roles for the kinase beyond biomineralization, including lipid homeostasis, wound healing, and cell migration and adhesion. Our results establish Fam20C as the major secretory pathway protein kinase and serve as a foundation for new areas of investigation into the role of secreted protein phosphorylation in human biology and disease.


Subject(s)
Casein Kinase I/chemistry , Casein Kinase I/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Amino Acid Sequence , Blood Proteins/metabolism , Casein Kinase I/genetics , Cell Adhesion , Cell Movement , Cerebrospinal Fluid Proteins/metabolism , Extracellular Matrix Proteins/genetics , Gene Knockout Techniques , Gene Ontology , Humans , Molecular Sequence Data , Phosphoproteins/analysis , Secretory Pathway , Substrate Specificity
5.
Mol Cell ; 81(21): 4527-4539.e8, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34407442

ABSTRACT

The kinase domain transfers phosphate from ATP to substrates. However, the Legionella effector SidJ adopts a kinase fold, yet catalyzes calmodulin (CaM)-dependent glutamylation to inactivate the SidE ubiquitin ligases. The structural and mechanistic basis in which the kinase domain catalyzes protein glutamylation is unknown. Here we present cryo-EM reconstructions of SidJ:CaM:SidE reaction intermediate complexes. We show that the kinase-like active site of SidJ adenylates an active-site Glu in SidE, resulting in the formation of a stable reaction intermediate complex. An insertion in the catalytic loop of the kinase domain positions the donor Glu near the acyl-adenylate for peptide bond formation. Our structural analysis led us to discover that the SidJ paralog SdjA is a glutamylase that differentially regulates the SidE ligases during Legionella infection. Our results uncover the structural and mechanistic basis in which the kinase fold catalyzes non-ribosomal amino acid ligations and reveal an unappreciated level of SidE-family regulation.


Subject(s)
Bacterial Proteins/chemistry , Protein Folding , Proteins/chemistry , Virulence Factors/chemistry , Bacterial Proteins/metabolism , Calmodulin/chemistry , Catalysis , Catalytic Domain , Cryoelectron Microscopy , Legionella/enzymology , Mutagenesis , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Domains , Spectrometry, Fluorescence , Ubiquitin-Protein Ligases/chemistry , Virulence Factors/metabolism
6.
Nature ; 609(7928): 793-800, 2022 09.
Article in English | MEDLINE | ID: mdl-35944563

ABSTRACT

The RNA genome of SARS-CoV-2 contains a 5' cap that facilitates the translation of viral proteins, protection from exonucleases and evasion of the host immune response1-4. How this cap is made in SARS-CoV-2 is not completely understood. Here we reconstitute the N7- and 2'-O-methylated SARS-CoV-2 RNA cap (7MeGpppA2'-O-Me) using virally encoded non-structural proteins (nsps). We show that the kinase-like nidovirus RdRp-associated nucleotidyltransferase (NiRAN) domain5 of nsp12 transfers the RNA to the amino terminus of nsp9, forming a covalent RNA-protein intermediate (a process termed RNAylation). Subsequently, the NiRAN domain transfers the RNA to GDP, forming the core cap structure GpppA-RNA. The nsp146 and nsp167 methyltransferases then add methyl groups to form functional cap structures. Structural analyses of the replication-transcription complex bound to nsp9 identified key interactions that mediate the capping reaction. Furthermore, we demonstrate in a reverse genetics system8 that the N terminus of nsp9 and the kinase-like active-site residues in the NiRAN domain are required for successful SARS-CoV-2 replication. Collectively, our results reveal an unconventional mechanism by which SARS-CoV-2 caps its RNA genome, thus exposing a new target in the development of antivirals to treat COVID-19.


Subject(s)
RNA Caps , RNA, Viral , SARS-CoV-2 , Viral Proteins , Antiviral Agents , COVID-19/virology , Catalytic Domain , Guanosine Diphosphate/metabolism , Humans , Methyltransferases/metabolism , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Protein Domains , RNA Caps/chemistry , RNA Caps/genetics , RNA Caps/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/enzymology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , COVID-19 Drug Treatment
7.
Proc Natl Acad Sci U S A ; 121(4): e2315925121, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38227654

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common type of soft tissue sarcoma in children and adolescents. Fusion-negative RMS (FN-RMS) accounts for more than 80% of all RMS cases. The long-term event-free survival rate for patients with high-grade FN-RMS is below 30%, highlighting the need for improved therapeutic strategies. CD73 is a 5' ectonucleotidase that hydrolyzes AMP to adenosine and regulates the purinergic signaling pathway. We found that CD73 is elevated in FN-RMS tumors that express high levels of TWIST2. While high expression of CD73 contributes to the pathogenesis of multiple cancers, its role in FN-RMS has not been investigated. We found that CD73 knockdown decreased FN-RMS cell growth while up-regulating the myogenic differentiation program. Moreover, mutation of the catalytic residues of CD73 rendered the protein enzymatically inactive and abolished its ability to stimulate FN-RMS growth. Overexpression of wildtype CD73, but not the catalytically inactive mutant, in CD73 knockdown FN-RMS cells restored their growth capacity. Likewise, treatment with an adenosine receptor A2A-B agonist partially rescued FN-RMS cell proliferation and bypassed the CD73 knockdown defective growth phenotype. These results demonstrate that the catalytic activity of CD73 contributes to the pathogenic growth of FN-RMS through the activation of the purinergic signaling pathway. Therefore, targeting CD73 and the purinergic signaling pathway represents a potential therapeutic approach for FN-RMS patients.


Subject(s)
Rhabdomyosarcoma , Adolescent , Child , Humans , Cell Differentiation/genetics , Cell Line, Tumor , Receptors, Purinergic P1 , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Signal Transduction
8.
Nature ; 542(7640): 197-202, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28114302

ABSTRACT

MicroRNAs (miRNAs) perform critical functions in normal physiology and disease by associating with Argonaute proteins and downregulating partially complementary messenger RNAs (mRNAs). Here we use clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) genome-wide loss-of-function screening coupled with a fluorescent reporter of miRNA activity in human cells to identify new regulators of the miRNA pathway. By using iterative rounds of screening, we reveal a novel mechanism whereby target engagement by Argonaute 2 (AGO2) triggers its hierarchical, multi-site phosphorylation by CSNK1A1 on a set of highly conserved residues (S824-S834), followed by rapid dephosphorylation by the ANKRD52-PPP6C phosphatase complex. Although genetic and biochemical studies demonstrate that AGO2 phosphorylation on these residues inhibits target mRNA binding, inactivation of this phosphorylation cycle globally impairs miRNA-mediated silencing. Analysis of the transcriptome-wide binding profile of non-phosphorylatable AGO2 reveals a pronounced expansion of the target repertoire bound at steady-state, effectively reducing the active pool of AGO2 on a per-target basis. These findings support a model in which an AGO2 phosphorylation cycle stimulated by target engagement regulates miRNA:target interactions to maintain the global efficiency of miRNA-mediated silencing.


Subject(s)
Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Gene Silencing , MicroRNAs/genetics , Amino Acid Sequence , Argonaute Proteins/chemistry , CRISPR-Cas Systems/genetics , Casein Kinase II/metabolism , HCT116 Cells , Humans , MicroRNAs/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity
9.
J Biol Chem ; 296: 100301, 2021.
Article in English | MEDLINE | ID: mdl-33476647

ABSTRACT

ADP-ribosyltransferases (ARTs) are a widespread superfamily of enzymes frequently employed in pathogenic strategies of bacteria. Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaire's disease, has acquired over 330 translocated effectors that showcase remarkable biochemical and structural diversity. However, the ART effectors that influence L. pneumophila have not been well defined. Here, we took a bioinformatic approach to search the Legionella effector repertoire for additional divergent members of the ART superfamily and identified an ART domain in Legionella pneumophila gene0181, which we hereafter refer to as Legionella ADP-Ribosyltransferase 1 (Lart1) (Legionella ART 1). We show that L. pneumophila Lart1 targets a specific class of 120-kDa NAD+-dependent glutamate dehydrogenase (GDH) enzymes found in fungi and protists, including many natural hosts of Legionella. Lart1 targets a conserved arginine residue in the NAD+-binding pocket of GDH, thereby blocking oxidative deamination of glutamate. Therefore, Lart1 could be the first example of a Legionella effector which directly targets a host metabolic enzyme during infection.


Subject(s)
ADP Ribose Transferases/chemistry , Bacterial Proteins/chemistry , Glutamate Dehydrogenase/chemistry , Legionella pneumophila/genetics , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , ADP-Ribosylation , Amino Acid Sequence , Amoeba/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Deamination , Escherichia coli/genetics , Escherichia coli/metabolism , Fungi , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Host-Pathogen Interactions , Kinetics , Legionella pneumophila/enzymology , Legionella pneumophila/pathogenicity , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
10.
J Biol Chem ; 295(18): 6214-6224, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32229585

ABSTRACT

The transfer of a phosphate from ATP to a protein substrate, a modification known as protein phosphorylation, is catalyzed by protein kinases. Protein kinases play a crucial role in virtually every cellular activity. Recent studies of atypical protein kinases have highlighted the structural similarity of the kinase superfamily despite notable differences in primary amino acid sequence. Here, using a bioinformatics screen, we searched for putative protein kinases in the intracellular bacterial pathogen Legionella pneumophila and identified the type 4 secretion system effector Lpg2603 as a remote member of the protein kinase superfamily. Employing an array of biochemical and structural biology approaches, including in vitro kinase assays and isothermal titration calorimetry, we show that Lpg2603 is an active protein kinase with several atypical structural features. Importantly, we found that the eukaryote-specific host signaling molecule inositol hexakisphosphate (IP6) is required for Lpg2603 kinase activity. Crystal structures of Lpg2603 in the apo-form and when bound to IP6 revealed an active-site rearrangement that allows for ATP binding and catalysis. Our results on the structure and activity of Lpg2603 reveal a unique mode of regulation of a protein kinase, provide the first example of a bacterial kinase that requires IP6 for its activation, and may aid future work on the function of this effector during Legionella pathogenesis.


Subject(s)
Host-Pathogen Interactions , Legionella pneumophila/enzymology , Phytic Acid/pharmacology , Protein Kinases/metabolism , Enzyme Activation/drug effects , Legionella pneumophila/physiology , Models, Molecular , Protein Conformation , Protein Kinases/chemistry
11.
Arterioscler Thromb Vasc Biol ; 39(10): 1996-2013, 2019 10.
Article in English | MEDLINE | ID: mdl-31553664

ABSTRACT

OBJECTIVE: PCSK9 (proprotein convertase subtilisin-kexin 9) enhances the degradation of the LDLR (low-density lipoprotein receptor) in endosomes/lysosomes. This study aimed to determine the sites of PCSK9 phosphorylation at Ser-residues and the consequences of such posttranslational modification on the secretion and activity of PCSK9 on the LDLR. Approach and Results: Fam20C (family with sequence similarity 20, member C) phosphorylates serines in secretory proteins containing the motif S-X-E/phospho-Ser, including the cholesterol-regulating PCSK9. In situ hybridization of Fam20C mRNA during development and in adult mice revealed a wide tissue distribution, including liver, but not small intestine. Here, we show that Fam20C phosphorylates PCSK9 at Serines 47, 666, 668, and 688. In hepatocytes, phosphorylation enhances PCSK9 secretion and maximizes its induced degradation of the LDLR via the extracellular and intracellular pathways. Replacing any of the 4 Ser by the phosphomimetic Glu or Asp enhanced PCSK9 activity only when the other sites are phosphorylated, whereas Ala substitutions reduced it, as evidenced by Western blotting, Elisa, and LDLR-immunolabeling. This newly uncovered PCSK9/LDLR regulation mechanism refines our understanding of the implication of global PCSK9 phosphorylation in the modulation of LDL-cholesterol and rationalizes the consequence of natural mutations, for example, S668R and E670G. Finally, the relationship of Ser-phosphorylation to the implication of PCSK9 in regulating LDL-cholesterol in the neurological Fragile X-syndrome disorder was investigated. CONCLUSIONS: Ser-phosphorylation of PCSK9 maximizes both its secretion and activity on the LDLR. Mass spectrometric approaches to measure such modifications were developed and applied to quantify the levels of bioactive PCSK9 in human plasma under normal and pathological conditions.


Subject(s)
Calcium-Binding Proteins/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Hyperlipoproteinemia Type II/genetics , Proprotein Convertase 9/metabolism , Receptors, LDL/genetics , Animals , Blotting, Western , Cells, Cultured , Hep G2 Cells , Hepatocytes/metabolism , Humans , Hyperlipoproteinemia Type II/physiopathology , In Situ Hybridization/methods , Male , Mice , Mice, Knockout , Microscopy, Confocal , Phosphorylation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/methods , Receptors, LDL/metabolism , Sensitivity and Specificity
12.
IUBMB Life ; 71(6): 749-759, 2019 06.
Article in English | MEDLINE | ID: mdl-30941842

ABSTRACT

Previous decades have seen an explosion in our understanding of protein kinase function in human health and disease. Hundreds of unique kinase structures have been solved, allowing us to create generalized rules for catalysis, assign roles of communities within the catalytic core, and develop specific drugs for targeting various pathways. Although our understanding of intracellular kinases has developed at a fast rate, our exploration into extracellular kinases has just begun. In this review, we will cover the secreted protein kinase families found in humans, bacteria, and parasites. © 2019 IUBMB Life, 71(6):749-759, 2019.


Subject(s)
Biological Transport/genetics , Phosphorylation/genetics , Protein Kinases/genetics , Animals , Bacteria/enzymology , Humans , Mammals/genetics , Parasites/enzymology , Protein Kinases/classification , Substrate Specificity
13.
Proc Natl Acad Sci U S A ; 113(25): E3482-91, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27185916

ABSTRACT

The modification of proteins by phosphorylation occurs in all life forms and is catalyzed by a large superfamily of enzymes known as protein kinases. We recently discovered a family of secretory pathway kinases that phosphorylate extracellular proteins. One member, family with sequence similarity 20C (Fam20C), is the physiological Golgi casein kinase. While examining distantly related protein sequences, we observed low levels of identity between the spore coat protein H (CotH), and the Fam20C-related secretory pathway kinases. CotH is a component of the spore in many bacterial and eukaryotic species, and is required for efficient germination of spores in Bacillus subtilis; however, the mechanism by which CotH affects germination is unclear. Here, we show that CotH is a protein kinase. The crystal structure of CotH reveals an atypical protein kinase-like fold with a unique mode of ATP binding. Examination of the genes neighboring cotH in B. subtilis led us to identify two spore coat proteins, CotB and CotG, as CotH substrates. Furthermore, we show that CotH-dependent phosphorylation of CotB and CotG is required for the efficient germination of B. subtilis spores. Collectively, our results define a family of atypical protein kinases and reveal an unexpected role for protein phosphorylation in spore biology.


Subject(s)
Protein Kinases , Spores, Bacterial/genetics , Bacillus subtilis/genetics , Bacterial Proteins/chemistry , Capsid Proteins , Phosphorylation
14.
Trends Biochem Sci ; 38(3): 121-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23276407

ABSTRACT

Protein kinases constitute one of the largest gene families and control many aspects of cellular life. In retrospect, the first indication for their existence was reported 130 years ago when the secreted protein, casein, was shown to contain phosphate. Despite its identification as the first phosphoprotein, the responsible kinase has remained obscure. This conundrum was solved with the discovery of a novel family of atypical protein kinases that are secreted and appear to phosphorylate numerous extracellular proteins, including casein. Fam20C, the archetypical member, phosphorylates secreted proteins within Ser-x-Glu/pSer motifs. This discovery has solved a 130-year-old mystery and has shed light on several human disorders of biomineralization.


Subject(s)
Protein Kinases/metabolism , Animals , Humans , Protein Kinases/genetics
15.
Proc Natl Acad Sci U S A ; 111(15): 5520-5, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24706917

ABSTRACT

The family with sequence similarity 20, member C (Fam20C) has recently been identified as the Golgi casein kinase. Fam20C phosphorylates secreted proteins on Ser-x-Glu/pSer motifs and loss-of-function mutations in the kinase cause Raine syndrome, an often-fatal osteosclerotic bone dysplasia. Fam20C is potentially an upstream regulator of the phosphate-regulating hormone fibroblast growth factor 23 (FGF23), because humans with FAM20C mutations and Fam20C KO mice develop hypophosphatemia due to an increase in full-length, biologically active FGF23. However, the mechanism by which Fam20C regulates FGF23 is unknown. Here we show that Fam20C directly phosphorylates FGF23 on Ser(180), within the FGF23 R(176)XXR(179)/S(180)AE subtilisin-like proprotein convertase motif. This phosphorylation event inhibits O-glycosylation of FGF23 by polypeptide N-acetylgalactosaminyltransferase 3 (GalNAc-T3), and promotes FGF23 cleavage and inactivation by the subtilisin-like proprotein convertase furin. Collectively, our results provide a molecular mechanism by which FGF23 is dynamically regulated by phosphorylation, glycosylation, and proteolysis. Furthermore, our findings suggest that cross-talk between phosphorylation and O-glycosylation of proteins in the secretory pathway may be an important mechanism by which secreted proteins are regulated.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factors/metabolism , Furin/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Amino Acid Sequence , Base Sequence , Casein Kinase I , DNA Primers/genetics , Extracellular Matrix Proteins/immunology , Fibroblast Growth Factor-23 , Glycosylation , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Molecular Sequence Data , Phosphorylation , Proteolysis , Sequence Analysis, DNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Polypeptide N-acetylgalactosaminyltransferase
16.
J Biol Chem ; 290(37): 22686-98, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26216881

ABSTRACT

Glycogen, the repository of glucose in many cell types, contains small amounts of covalent phosphate, of uncertain function and poorly understood metabolism. Loss-of-function mutations in the laforin gene cause the fatal neurodegenerative disorder, Lafora disease, characterized by increased glycogen phosphorylation and the formation of abnormal deposits of glycogen-like material called Lafora bodies. It is generally accepted that the phosphate is removed by the laforin phosphatase. To study the dynamics of skeletal muscle glycogen phosphorylation in vivo under physiological conditions, mice were subjected to glycogen-depleting exercise and then monitored while they resynthesized glycogen. Depletion of glycogen by exercise was associated with a substantial reduction in total glycogen phosphate and the newly resynthesized glycogen was less branched and less phosphorylated. Branching returned to normal on a time frame of days, whereas phosphorylation remained suppressed over a longer period of time. We observed no change in markers of autophagy. Exercise of 3-month-old laforin knock-out mice caused a similar depletion of glycogen but no loss of glycogen phosphate. Furthermore, remodeling of glycogen to restore the basal branching pattern was delayed in the knock-out animals. From these results, we infer that 1) laforin is responsible for glycogen dephosphorylation during exercise and acts during the cytosolic degradation of glycogen, 2) excess glycogen phosphorylation in the absence of laforin delays the normal remodeling of the branching structure, and 3) the accumulation of glycogen phosphate is a relatively slow process involving multiple cycles of glycogen synthesis-degradation, consistent with the slow onset of the symptoms of Lafora disease.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Glycogen/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Animals , Dual-Specificity Phosphatases/genetics , Glycogen/genetics , Lafora Disease/genetics , Lafora Disease/metabolism , Lafora Disease/pathology , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Phosphorylation/genetics , Protein Tyrosine Phosphatases, Non-Receptor
17.
Biochim Biophys Acta ; 1854(10 Pt B): 1687-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25862977

ABSTRACT

Protein phosphorylation is a nearly universal post-translation modification involved in a plethora of cellular events. Even though phosphorylation of extracellular proteins had been observed, the identity of the kinases that phosphorylate secreted proteins remained a mystery until only recently. Advances in genome sequencing and genetic studies have paved the way for the discovery of a new class of kinases that localize within the endoplasmic reticulum, Golgi apparatus and the extracellular space. These novel kinases phosphorylate proteins and proteoglycans in the secretory pathway and appear to regulate various extracellular processes. Mutations in these kinases cause human disease, thus underscoring the biological importance of phosphorylation within the secretory pathway. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Subject(s)
Abnormalities, Multiple/genetics , Casein Kinase I/genetics , Cleft Palate/genetics , Exophthalmos/genetics , Extracellular Matrix Proteins/genetics , Microcephaly/genetics , Osteosclerosis/genetics , Protein-Tyrosine Kinases/genetics , Secretory Pathway/genetics , Abnormalities, Multiple/enzymology , Animals , Casein Kinase I/chemistry , Casein Kinase I/metabolism , Cleft Palate/enzymology , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Exophthalmos/enzymology , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , Humans , Microcephaly/enzymology , Mutation , Osteosclerosis/enzymology , Phosphorylation/genetics , Protein Conformation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Substrate Specificity
18.
Biochim Biophys Acta ; 1854(10 Pt B): 1718-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25936777

ABSTRACT

Fam20C is an atypical kinase implicated in bio-mineralization and phosphate homeostasis disorders, and has recently been shown to account for the activity of an orphan enzyme ("genuine casein kinase", G-CK) previously characterized for its ability to phosphorylate casein and a plethora of secreted proteins at serine residues specified by the S-x-E/pS motif. Fam20C/G-CK activity is only appreciable in the presence of high Mn2+ concentration (>1 mM), and is negligible if Mn2+ is replaced by physiological Mg2+ concentrations. Here we show that sphingosine (but not its biological precursor ceramide) not only stimulates several-fold Fam20C activity in the presence of Mn2+, but also confers a comparable activity to Fam20C assayed with Mg2+. Activation by sphingosine is evident using a variety of substrates, and is accounted for by both higher Vmax and decreased KmATP, as judged from kinetics run with the ß(28-40) substrate peptide and a physiological substrate, BMP-15. Sphingosine also protects Fam20C from thermal inactivation. Consistent with the in vitro results, by treating Fam20C expressing HEK293T cells with myriocin, a potent inhibitor of the sphingosine biosynthetic pathway, the activity of Fam20C released into the conditioned medium is substantially decreased corroborating the concept that sphingosine (or related metabolite(s)) is a co-factor required by Fam20C to optimally display its biological functions. None of the small molecule kinase inhibitors tested so far were able to inhibit Fam20C. Interestingly however fingolimod, an immunosuppressive drug structurally related to sphingosine, used for the treatment of multiple sclerosis, is a powerful activator of Fam20C, both wild type and its pathogenic, loss of function, T268M mutant. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Subject(s)
Casein Kinase I/genetics , Extracellular Matrix Proteins/genetics , Multiple Sclerosis/genetics , Sphingosine/biosynthesis , Amino Acid Sequence , Casein Kinase I/chemistry , Extracellular Matrix Proteins/chemistry , Fatty Acids, Monounsaturated/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Multiple Sclerosis/enzymology , Phosphorylation , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , Transcriptional Activation/drug effects
19.
Proc Natl Acad Sci U S A ; 110(26): 10574-9, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23754375

ABSTRACT

The family with sequence similarity 20 (Fam20) kinases phosphorylate extracellular substrates and play important roles in biomineralization. Fam20C is the Golgi casein kinase that phosphorylates secretory pathway proteins within Ser-x-Glu/pSer motifs. Mutations in Fam20C cause Raine syndrome, an osteosclerotic bone dysplasia. Here we report the crystal structure of the Fam20C ortholog from Caenorhabditis elegans. The nucleotide-free and Mn/ADP-bound structures unveil an atypical protein kinase-like fold and highlight residues critical for activity. The position of the regulatory αC helix and the lack of an activation loop indicate an architecture primed for efficient catalysis. Furthermore, several distinct elements, including the presence of disulfide bonds, suggest that the Fam20 family diverged early in the evolution of the protein kinase superfamily. Our results reinforce the structural diversity of protein kinases and have important implications for patients with disorders of biomineralization.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Casein Kinases/chemistry , Amino Acid Sequence , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Casein Kinase I , Casein Kinases/genetics , Casein Kinases/metabolism , Crystallography, X-Ray , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Golgi Apparatus/enzymology , Humans , Models, Molecular , Molecular Sequence Data , Sequence Homology, Amino Acid , Substrate Specificity
20.
Proc Natl Acad Sci U S A ; 110(52): 20976-81, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324135

ABSTRACT

Glycogen is a glucose polymer that contains minor amounts of covalently attached phosphate. Hyperphosphorylation is deleterious to glycogen structure and can lead to Lafora disease. Recently, it was demonstrated that glycogen synthase catalyzes glucose-phosphate transfer in addition to its characteristic glucose transfer reaction. Glucose-1,2-cyclic-phosphate (GCP) was proposed to be formed from UDP-Glc breakdown and subsequently transferred, thus providing a source of phosphate found in glycogen. To gain further insight into the molecular basis for glucose-phosphate transfer, two structures of yeast glycogen synthase were determined; a 3.0-Å resolution structure of the complex with UMP/GCP and a 2.8-Å resolution structure of the complex with UDP/glucose. Structural superposition of the complexes revealed that the bound ligands and most active site residues are positioned similarly, consistent with the use of a common transfer mechanism for both reactions. The N-terminal domain of the UDP-glucose complex was found to be 13.3° more closed compared with a UDP complex. However, the UMP · GCP complex was 4.8° less closed than the glucose complex, which may explain the low efficiency of GCP transfer. Modeling of either α- or ß-glucose or a mixture of both anomers can account for the observed electron density of the UDP-glucose complex. NMR studies of UDP-Glc hydrolysis by yeast glycogen synthase were used to verify the stereochemistry of the product, and they also showed synchronous GCP accumulation. The similarities in the active sites of glycogen synthase and glycogen phosphorylase support the idea of a common catalytic mechanism in GT-B enzymes independent of the specific reaction catalyzed.


Subject(s)
Glycogen Synthase/metabolism , Glycogen/chemistry , Models, Molecular , Phosphates/chemistry , Crystallography , Glycogen/metabolism , Glycogen Synthase/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mutagenesis , Phosphates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL