Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 430
Filter
1.
Nature ; 627(8003): 431-436, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383786

ABSTRACT

To survive bacteriophage (phage) infections, bacteria developed numerous anti-phage defence systems1-7. Some of them (for example, type III CRISPR-Cas, CBASS, Pycsar and Thoeris) consist of two modules: a sensor responsible for infection recognition and an effector that stops viral replication by destroying key cellular components8-12. In the Thoeris system, a Toll/interleukin-1 receptor (TIR)-domain protein, ThsB, acts as a sensor that synthesizes an isomer of cyclic ADP ribose, 1''-3' glycocyclic ADP ribose (gcADPR), which is bound in the Smf/DprA-LOG (SLOG) domain of the ThsA effector and activates the silent information regulator 2 (SIR2)-domain-mediated hydrolysis of a key cell metabolite, NAD+ (refs. 12-14). Although the structure of ThsA has been solved15, the ThsA activation mechanism remained incompletely understood. Here we show that 1''-3' gcADPR, synthesized in vitro by the dimeric ThsB' protein, binds to the ThsA SLOG domain, thereby activating ThsA by triggering helical filament assembly of ThsA tetramers. The cryogenic electron microscopy (cryo-EM) structure of activated ThsA revealed that filament assembly stabilizes the active conformation of the ThsA SIR2 domain, enabling rapid NAD+ depletion. Furthermore, we demonstrate that filament formation enables a switch-like response of ThsA to the 1''-3' gcADPR signal.


Subject(s)
Bacteria , Bacterial Proteins , Bacteriophages , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/biosynthesis , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Bacteria/metabolism , Bacteria/virology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Bacteriophages/chemistry , Bacteriophages/metabolism , Bacteriophages/ultrastructure , Cryoelectron Microscopy , Hydrolysis , NAD/metabolism , Protein Domains , Protein Multimerization , Protein Stability
2.
J Org Chem ; 84(10): 6143-6157, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30978018

ABSTRACT

TRPM2 (transient receptor potential cation channel, subfamily M, member 2) is a nonselective cation channel involved in the response to oxidative stress and in inflammation. Its role in autoimmune and neurodegenerative diseases makes it an attractive pharmacological target. Binding of the nucleotide adenosine 5'-diphosphate ribose (ADPR) to the cytosolic NUDT9 homology (NUDT9 H) domain activates the channel. A detailed understanding of how ADPR interacts with the TRPM2 ligand binding domain is lacking, hampering the rational design of modulators, but the terminal ribose of ADPR is known to be essential for activation. To study its role in more detail, we designed synthetic routes to novel analogues of ADPR and 2'-deoxy-ADPR that were modified only by removal of a single hydroxyl group from the terminal ribose. The ADPR analogues were obtained by coupling nucleoside phosphorimidazolides to deoxysugar phosphates. The corresponding C2″-based analogues proved to be unstable. The C1″- and C3″-ADPR analogues were evaluated electrophysiologically by patch-clamp in TRPM2-expressing HEK293 cells. In addition, a compound with all hydroxyl groups of the terminal ribose blocked as its 1″-ß- O-methyl-2″,3″- O-isopropylidene derivative was evaluated. Removal of either C1″ or C3″ hydroxyl groups from ADPR resulted in loss of agonist activity. Both these modifications and blocking all three hydroxyl groups resulted in TRPM2 antagonists. Our results demonstrate the critical role of these hydroxyl groups in channel activation.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Molecular Probes/chemical synthesis , Molecular Probes/metabolism , TRPM Cation Channels/metabolism , Chemistry Techniques, Synthetic , HEK293 Cells , Humans , Models, Molecular , Molecular Probes/chemistry , Protein Conformation , TRPM Cation Channels/chemistry
3.
Nat Commun ; 10(1): 218, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30644400

ABSTRACT

Tpt1 is an essential agent of fungal tRNA splicing that removes the 2'-PO4 at the splice junction generated by fungal tRNA ligase. Tpt1 catalyzes a unique two-step reaction whereby the 2'-PO4 attacks NAD+ to form an RNA-2'-phospho-ADP-ribosyl intermediate that undergoes transesterification to yield 2'-OH RNA and ADP-ribose-1″,2″-cyclic phosphate products. Because Tpt1 is inessential in exemplary bacterial and mammalian taxa, Tpt1 is seen as an attractive antifungal target. Here we report a 1.4 Šcrystal structure of Tpt1 in a product-mimetic complex with ADP-ribose-1″-phosphate in the NAD+ site and pAp in the RNA site. The structure reveals how Tpt1 recognizes a 2'-PO4 RNA splice junction and the mechanism of RNA phospho-ADP-ribosylation. This study also provides evidence that a bacterium has an endogenous phosphorylated substrate with which Tpt1 reacts.


Subject(s)
Bacterial Proteins/metabolism , Clostridium thermocellum/enzymology , RNA, Transfer/metabolism , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Ligands , NAD/metabolism , Phosphates/metabolism , Protein Conformation
4.
PLoS One ; 13(11): e0207693, 2018.
Article in English | MEDLINE | ID: mdl-30500844

ABSTRACT

Cholera toxin, an 84-kDa multimeric protein and a major virulence factor of Vibrio cholerae, uses the ADP-ribosyltransferase activity of its A subunit to intoxicate host cells. ADP-ribosylation is a posttranslational modification of proteins, in which the ADP-ribose moiety of NAD+ is transferred to an acceptor. In mammalian cells, ADP-ribosylation of acceptors appears to be reversible. ADP-ribosyltransferases (ARTs) catalyze the modification of acceptor proteins, and ADP-ribose-acceptor hydrolases (ARHs) cleave the ADP-ribose-acceptor bond. ARH1 specifically cleaves the ADP-ribose-arginine bond. We previously demonstrated a role for endogenous ARH1 in regulating the extent of cholera toxin-mediated fluid and electrolyte abnormalities in a mouse model of intoxication. Murine ARH1-knockout (KO) cells and ARH1-KO mice exhibited increased sensitivity to cholera toxin compared to their wild-type (WT) counterparts. In the current report, we examined the sensitivity to cholera toxin of male and female ARH1-KO and WT mice. Intestinal loops derived from female ARH1-KO mice when injected with cholera toxin showed increased fluid accumulation compared to male ARH1-KO mice. WT mice did not show gender differences in fluid accumulation, ADP-ribosylarginine content, and ADP-ribosyl Gαs levels. Injection of 8-Bromo-cAMP into the intestinal loops also increased fluid accumulation, however, there was no significant difference between female and male mice or in WT and KO mice. Female ARH1-KO mice showed greater amounts of ADP-ribosylated Gαs protein and increased ADP-ribosylarginine content both in whole intestine and in epithelial cells than did male ARH1-KO mice. These results demonstrate that female ARH1-KO mice are more sensitive to cholera toxin than male mice. Loss of ARH1 confers gender sensitivity to the effects of cholera toxin but not of cyclic AMP. These observations may in part explain the finding noted in some clinical reports of enhanced symptoms of cholera and/or diarrhea in women than men.


Subject(s)
Body Fluids/metabolism , Cholera Toxin/metabolism , Intestinal Mucosa/metabolism , N-Glycosyl Hydrolases/deficiency , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Animals , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Intestinal Mucosa/cytology , Male , Mice, Knockout , N-Glycosyl Hydrolases/genetics , Sex Factors , Vibrio cholerae/metabolism
5.
Methods Mol Biol ; 1813: 149-165, 2018.
Article in English | MEDLINE | ID: mdl-30097866

ABSTRACT

Methods are described for determination of arginine-specific mono-ADP-ribosyltransferase activity of purified proteins and intact cells by monitoring the transfer of ADP-ribose from NAD+ to a model substrate, e.g., arginine, agmatine, and peptide (human neutrophil peptide-1 [HNP1]), and for the nonenzymatic hydrolysis of ADP-ribose-arginine to ornithine, a noncoded amino acid. In addition, preparation of purified ADP-ribosylarginine is included as a control substrate for ADP-ribosylation reactions.


Subject(s)
ADP Ribose Transferases/isolation & purification , ADP-Ribosylation/genetics , Adenosine Diphosphate Ribose/isolation & purification , Molecular Biology/methods , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/genetics , Arginine/chemistry , Catalysis , Humans , Ornithine/chemistry
6.
Chem Biol Drug Des ; 91(2): 552-566, 2018 02.
Article in English | MEDLINE | ID: mdl-29034580

ABSTRACT

Transient receptor potential melastatin-2 (TRPM2) channel critical for monitoring internal body temperature is implicated in the pathological processes such as neurodegeneration. However, lacking selective and potent TRPM2 inhibitors impedes investigation and validation of the channel as a drug target. To discover novel and selective TRPM2 inhibitors, a series of adenosine 5'-diphosphoribose analogues were synthesized, and their activities and selectivity were evaluated. Whole-cell patch-clamp recordings were employed for screen and evaluation of synthesized compounds. Two compounds, 7i and 8a, were identified as TRPM2 inhibitors with IC50 of 5.7 and 5.4 µm, respectively. Both 7i and 8a inhibited TRPM2 current without affecting TRPM7, TRPM8, TRPV1 and TRPV3. These two TRPM2 inhibitors can serve as new pharmacological tools for further investigation and validation of TRPM2 channel as a drug target, and the summarized structure-activity relationship (SAR) may also provide insights into further improving existing inhibitors as potential lead compounds.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , TRPM Cation Channels/metabolism , Adenosine Diphosphate Ribose/chemical synthesis , Adenosine Diphosphate Ribose/metabolism , Calcium/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Patch-Clamp Techniques , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Structure-Activity Relationship , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
7.
Sci Rep ; 7(1): 12439, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963484

ABSTRACT

ADP-ribosylhydrolases (ARH1, ARH2 and ARH3) are a family of enzymes to catalyze ADP-ribosylation, a reversible and covalent post-translational modification (PTM). There are four phosphorylated sites (Tyr-4, Tyr-19, Tyr-20, and Tyr-205) in ARH1. To explore the structural changes and functional impact induced by phosphorylation, molecular dynamics (MD) simulations and steered molecular dynamics (SMD) simulations were performed for the phosphorylated and non-phosphorylated ARH1 with the ligands. MD simulations results indicate that: (1) Glu-25 is more frequently in the α helix group in the phosphorylated state with the adenosine-5-diphosphate-ribosylarginine (ADP-RA) complex (51.56%) than that of the non-phosphorylated state(2.12%); (2) Ser-124 and Ser-264 become less flexible in the phosphorylated state with ADP-RA complex, which helps two residues form hydrogen bonds with ADP-RA; and (3) Tyr-211 is also less flexible in the phosphorylated state with ADP-RA complex, which helps stabilize the cation-π interaction of Y211-R119. All these changes facilitate ADP-RA to bind ARH1. In addition, according to the crystal structure of adenosine-5-diphosphate-ribose (ADP-ribose) in complex with non-phosphorylated and phosphorylated ARH1, the possible unbinding pathways of ADP-ribose from non-phosphorylated and phosphorylated ARH1 were explored respectively using SMD simulations. Our results show that phosphorylated ARH1 has more ordered structures than the non-phosphorylated type.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemistry , Molecular Dynamics Simulation , Protein Processing, Post-Translational , ADP-Ribosylation , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Diphosphate Ribose/metabolism , Binding Sites , Humans , Kinetics , Ligands , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Substrate Specificity , Thermodynamics
8.
Cell Chem Biol ; 24(8): 919-920, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28820960

ABSTRACT

This month: Lysosomal iron linked to cell death in cancer stem cells, non-enzymatic catalyst SynCAc for histone acylation, cytotoxins ivermectin and etoposide bring new anti-fungals out of the crypt, and 2'-deoxy-ADPR as second messenger activating TRPM2.


Subject(s)
Histones/metabolism , Iron/metabolism , Second Messenger Systems/physiology , Acylation , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Humans , Multigene Family , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , TRPM Cation Channels/chemistry , TRPM Cation Channels/metabolism
9.
Nat Chem Biol ; 13(9): 1036-1044, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28671679

ABSTRACT

Transient receptor potential melastatin 2 (TRPM2) is a ligand-gated Ca2+-permeable nonselective cation channel. Whereas physiological stimuli, such as chemotactic agents, evoke controlled Ca2+ signals via TRPM2, pathophysiological stimuli such as reactive oxygen species and genotoxic stress result in prolonged TRPM2-mediated Ca2+ entry and, consequently, apoptosis. To date, adenosine 5'-diphosphoribose (ADPR) has been assumed to be the main agonist for TRPM2. Here we show that 2'-deoxy-ADPR was a significantly better TRPM2 agonist, inducing 10.4-fold higher whole-cell currents at saturation. Mechanistically, this increased activity was caused by a decreased rate of inactivation and higher average open probability. Using high-performance liquid chromatography (HPLC) and mass spectrometry, we detected endogenous 2'-deoxy-ADPR in Jurkat T lymphocytes. Consistently, cytosolic nicotinamide mononucleotide adenylyltransferase 2 (NMNAT-2) and nicotinamide adenine dinucleotide (NAD)-glycohydrolase CD38 sequentially catalyzed the synthesis of 2'-deoxy-ADPR from nicotinamide mononucleotide (NMN) and 2'-deoxy-ATP in vitro. Thus, 2'-deoxy-ADPR is an endogenous TRPM2 superagonist that may act as a cell signaling molecule.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Clusterin/agonists , ADP-ribosyl Cyclase 1/chemistry , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/pharmacology , Chromatography, High Pressure Liquid , Humans , Hydrogen Peroxide/chemistry , Jurkat Cells , Molecular Structure , Signal Transduction/drug effects
10.
Chembiochem ; 18(16): 1616-1626, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28589630

ABSTRACT

The design of a bioreversibly protected lipophilic sugar nucleotide as a potential membrane-permeable precursor of adenosine diphosphate ribose (ADPR) is described. ADPR is the most potent activator of the transient receptor potential melastatin 2 (TRPM2) ion channel. Membrane-permeable, lipophilic derivatives of ADPR are of great interest as tools for study of the mechanism of TRPM2. The approach described here was based on our recently disclosed "DiPPro" and "TriPPPro" prodrug approaches developed for the intracellular delivery of nucleotides. A lipophilic, bioreversibly masked ADPR analogue containing an enzymatically cleavable 4-pentanoyloxybenzyl (PB) mask at the phosphate moiety next to the 5'-position of adenosine, together with O-acetyl groups, was prepared in high yields. Chemical and enzymatic hydrolysis studies in phosphate buffer (pH 7.3) were performed to assess chemical stability and possible (selective) enzymatic demasking of the ADPR analogue. HPLC-MS revealed that the PB group was readily cleaved enzymatically. In addition, the formation of partially deacetylated ADPR compounds and also of fully unprotected ADPR was observed.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemical synthesis , Prodrugs/chemical synthesis , Adenosine Diphosphate Ribose/chemistry , Animals , Carboxylic Ester Hydrolases/chemistry , Drug Stability , Hydrogen-Ion Concentration , Hydrolysis , Prodrugs/chemistry , Swine
11.
Biochem Soc Trans ; 43(3): 417-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26009185

ABSTRACT

Synthetic compounds open up new avenues to interrogate and manipulate intracellular Ca2+ signalling pathways. They may ultimately lead to drug-like analogues to intervene in disease. Recent advances in chemical biology tools available to probe Ca2+ signalling are described, with a particular focus on those synthetic analogues from our group that have enhanced biological understanding or represent a step towards more drug-like molecules. Adenophostin (AdA) is the most potent known agonist at the inositol 1,4,5-trisphosphate receptor (IP3R) and synthetic analogues provide a binding model for receptor activation and channel opening. 2-O-Modified inositol 1,4,5-trisphosphate (IP3) derivatives that are partial agonists at the IP3R reveal key conformational changes of the receptor upon ligand binding. Biphenyl polyphosphates illustrate that simple non-inositol surrogates can be engineered to give prototype IP3R agonists or antagonists and act as templates for protein co-crystallization. Cyclic adenosine 5'-diphosphoribose (cADPR) can be selectively modified using total synthesis, generating chemically and biologically stable tools to investigate Ca2+ release via the ryanodine receptor (RyR) and to interfere with cADPR synthesis and degradation. The first neutral analogues with a synthetic pyrophosphate bioisostere surprisingly retain the ability to release Ca2+, suggesting a new route to membrane-permeant tools. Adenosine 5'-diphosphoribose (ADPR) activates the Ca2+-, Na+- and K+-permeable transient receptor potential melastatin 2 (TRPM2) cation channel. Synthetic ADPR analogues provide the first structure-activity relationship (SAR) for this emerging messenger and the first functional antagonists. An analogue based on the nicotinic acid motif of nicotinic acid adenine dinucleotide phosphate (NAADP) antagonizes NAADP-mediated Ca2+ release in vitro and is effective in vivo against induced heart arrhythmia and autoimmune disease, illustrating the therapeutic potential of targeted small molecules.


Subject(s)
Adenosine Diphosphate Ribose/chemistry , Arrhythmias, Cardiac/drug therapy , Calcium Signaling/drug effects , Calcium/metabolism , Small Molecule Libraries/therapeutic use , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/therapeutic use , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemical synthesis , Arrhythmias, Cardiac/pathology , Calcium Channel Blockers/therapeutic use , Humans , Inositol 1,4,5-Trisphosphate/genetics , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , NADP/analogs & derivatives , NADP/antagonists & inhibitors , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
12.
Biochem J ; 468(3): 475-84, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25849365

ABSTRACT

Although most sequenced members of the industrially important ketol-acid reductoisomerase (KARI) family are class I enzymes, structural studies to date have focused primarily on the class II KARIs, which arose through domain duplication. In the present study, we present five new crystal structures of class I KARIs. These include the first structure of a KARI with a six-residue ß2αB (cofactor specificity determining) loop and an NADPH phosphate-binding geometry distinct from that of the seven- and 12-residue loops. We also present the first structures of naturally occurring KARIs that utilize NADH as cofactor. These results show insertions in the specificity loops that confounded previous attempts to classify them according to loop length. Lastly, we explore the conformational changes that occur in class I KARIs upon binding of cofactor and metal ions. The class I KARI structures indicate that the active sites close upon binding NAD(P)H, similar to what is observed in the class II KARIs of rice and spinach and different from the opening of the active site observed in the class II KARI of Escherichia coli. This conformational change involves a decrease in the bending of the helix that runs between the domains and a rearrangement of the nicotinamide-binding site.


Subject(s)
Alicyclobacillus/enzymology , Azotobacter vinelandii/enzymology , Bacterial Proteins/metabolism , Coenzymes/metabolism , Desulfurococcaceae/enzymology , Ketol-Acid Reductoisomerase/metabolism , Models, Molecular , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalytic Domain , Coenzymes/chemistry , Crystallography, X-Ray , Ketol-Acid Reductoisomerase/chemistry , Ketol-Acid Reductoisomerase/genetics , Magnesium/chemistry , Magnesium/metabolism , Molecular Conformation , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , NAD/chemistry , NAD/metabolism , NADP/chemistry , NADP/metabolism , Phosphorylation , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment
13.
Sci Rep ; 5: 8959, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25753532

ABSTRACT

Control of immunologic tolerance and homeostasis rely on Foxp3(+)CD4(+)CD25(+) regulatory T cells (Tregs) that constitutively express the high affinity receptor for Interleukin-2, CD25. Tregs proliferate in response to injections of IL-2/anti-IL-2 antibody complexes or low doses of IL-2. However, little is known about endogenous mechanisms that regulate the sensitivity of CD25 to signaling by IL-2. Here we demonstrate that CD25 is ADP-ribosylated at Arg35 in the IL-2 binding site by ecto-ADP-ribosyltransferase ARTC2.2, a toxin-related GPI-anchored ecto-enzyme. ADP-ribosylation inhibits binding of IL-2 by CD25, IL-2- induced phosphorylation of STAT5, and IL-2-dependent cell proliferation. Our study elucidates an as-yet-unrecognized mechanism to tune IL-2 signaling. This newly found mechanism might thwart Tregs at sites of inflammation and thereby permit a more potent response of activated effector T cells.


Subject(s)
Immune Tolerance , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/metabolism , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/genetics , HEK293 Cells , Humans , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Phosphorylation , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology
14.
BMC Biochem ; 15: 26, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25494717

ABSTRACT

BACKGROUND: Cholix toxin is an ADP-ribosyltransferase found in non-O1/non-O139 strains of Vibrio cholera. The catalytic fragment of cholix toxin was characterized as a diphthamide dependent ADP-ribosyltransferase. RESULTS: Our studies on the enzymatic activity of cholix toxin catalytic fragment show that the transfer of ADP-ribose to toxin takes place by a predominantly intramolecular mechanism and results in the preferential alkylation of arginine residues proximal to the NAD+ binding pocket. Multiple arginine residues, located near the catalytic site and at distal sites, can be the ADP-ribose acceptor in the auto-reaction. Kinetic studies of a model enzyme, M8, showed that a diffusible intermediate preferentially reacted with arginine residues in proximity to the NAD+ binding pocket. ADP-ribosylarginine activity of cholix toxin catalytic fragment could also modify exogenous substrates. Auto-ADP-ribosylation of cholix toxin appears to have negatively regulatory effect on ADP-ribosylation of exogenous substrate. However, at the presence of both endogenous and exogenous substrates, ADP-ribosylation of exogenous substrates occurred more efficiently than that of endogenous substrates. CONCLUSIONS: We discovered an ADP-ribosylargininyl activity of cholix toxin catalytic fragment from our studies in auto-ADP-ribosylation, which is mediated through diffusible intermediates. The lifetime of the hypothetical intermediate exceeds recorded and predicted lifetimes for the cognate oxocarbenium ion. Therefore, a diffusible strained form of NAD+ intermediate was proposed to react with arginine residues in a proximity dependent manner.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Cholera Toxin/metabolism , Adenosine Diphosphate Ribose/toxicity , Diffusion
15.
Org Lett ; 15(9): 2306-9, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23614697

ABSTRACT

The glycosylation properties of ribofuranosyl N-phenyltrifluoroacetimidates toward carboxamide side chains of asparagine and glutamine were investigated. Conditions were found that promote nearly exclusive formation of the α-anomerically configured N-glycosides. The strategy allows for the synthesis of Fmoc-amino acids suitably modified for the preparation of ADP-ribosylated peptides. Furthermore, ribosylation of serine with these donors proved to be completely α-selective, and for the first time, α-ribosylated glutamic and aspartic acid, the naturally occurring sites for poly-ADP-ribosylation, were synthesized.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/chemistry , Amino Acids/chemical synthesis , Aspartic Acid/chemical synthesis , Glutamic Acid/chemical synthesis , Ribose/analogs & derivatives , Ribose/chemistry , Amino Acids/chemistry , Aspartic Acid/chemistry , Glutamic Acid/chemistry , Glycosylation , Molecular Structure , Stereoisomerism
16.
Cancer Res ; 71(15): 5327-35, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21697277

ABSTRACT

Protein ADP-ribosylation is a reversible posttranslational modification of uncertain significance in cancer. In this study, we evaluated the consequences for cancer susceptibility in the mouse of a genetic deletion of the enzyme responsible for removing mono-ADP-ribose moieties from arginines in cellular proteins. Specifically, we analyzed cancer susceptibility in animals lacking the ADP-ribosylarginine hydrolase (ARH1) that cleaves the ADP ribose-protein bond. ARH1(-/-) cells or ARH1(-/-) cells overexpressing an inactive mutant ARH1 protein (ARH1(-/-)+dm) had higher proliferation rates than either wild-type ARH1(+/+) cells or ARH1(-/-) cells engineered to express the wild-type ARH1 enzyme. More significantly, ARH1(-/-) and ARH1(+/-) mice spontaneously developed lymphomas, adenocarcinomas, and metastases more frequently than wild-type ARH1(+/+) mice. In ARH1(+/-) mice, we documented in all arising tumors mutation of the remaining wild-type allele (or loss of heterozygosity), illustrating the strict correlation that existed between tumor formation and absence of ARH1 gene function. Our findings show that proper control of protein ADP-ribosylation levels affected by ARH1 is essential for cancer suppression.


Subject(s)
Cell Transformation, Neoplastic/genetics , N-Glycosyl Hydrolases/physiology , Neoplasm Metastasis/genetics , Neoplasm Proteins/physiology , Neoplasms, Experimental/enzymology , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Animals , Cell Division , Cell Transformation, Neoplastic/metabolism , Female , Genetic Predisposition to Disease , Genotype , Loss of Heterozygosity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , N-Glycosyl Hydrolases/deficiency , N-Glycosyl Hydrolases/genetics , Neoplasms, Experimental/genetics , Protein Processing, Post-Translational , Tumor Stem Cell Assay
17.
Mol Cell Endocrinol ; 333(1): 8-19, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21094205

ABSTRACT

The mechanism by which extracellular ADP ribose (ADPr) increases intracellular free Ca(2+) concentration ([Ca(2+)](i)) remains unknown. We measured [Ca(2+)](i) changes in fura-2 loaded rat insulinoma INS-1E cells, and in primary ß-cells from rat and human. A phosphonate analogue of ADPr (PADPr) and 8-Bromo-ADPr (8Br-ADPr) were synthesized. ADPr increased [Ca(2+)](i) in the form of a peak followed by a plateau dependent on extracellular Ca(2+). NAD(+), cADPr, PADPr, 8Br-ADPr or breakdown products of ADPr did not increase [Ca(2+)](i). The ADPr-induced [Ca(2+)](i) increase was not affected by inhibitors of TRPM2, but was abolished by thapsigargin and inhibited when phospholipase C and IP(3) receptors were inhibited. MRS 2179 and MRS 2279, specific inhibitors of the purinergic receptor P2Y1, completely blocked the ADPr-induced [Ca(2+)](i) increase. ADPr increased [Ca(2+)](i) in transfected human astrocytoma cells (1321N1) that express human P2Y1 receptors, but not in untransfected astrocytoma cells. We conclude that ADPr is a specific agonist of P2Y1 receptors.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Purinergic P2Y Receptor Agonists/metabolism , Receptors, Purinergic P2Y1/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Calcium/metabolism , Cell Line, Tumor , Cells, Cultured , Halogenation , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Insulin-Secreting Cells/metabolism , Ligands , Purinergic P2Y Receptor Antagonists/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Thapsigargin/pharmacology , Type C Phospholipases/antagonists & inhibitors
18.
Naunyn Schmiedebergs Arch Pharmacol ; 380(6): 497-507, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19894037

ABSTRACT

A pharmacological characterization of Ca(2+) influx pathways in neutrophil granulocytes is problematic because of the lack of specific inhibitors. The activation of transient receptor potential cation channel, subfamily M, member 2 (TRPM2) channels by intracellular adenosine diphosphate ribose (ADPR), well characterized in neutrophils, is reportedly inhibited by 8-bromo-ADPR (8Br-ADPR). TRPM2 is blocked by N-(p-amylcinnamoyl)anthranilic acid (ACA) interfering with the pore, but ACA is as well effective on other transient receptor potential channels, especially transient receptor potential canonical (TRPC) channels. We wished to analyze whether ACA and 8Br-ADPR were suitable probes to demonstrate that different Ca(2+) entry pathways are activated in human neutrophil granulocytes by the receptor-dependent stimuli N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP) and platelet-activating factor (PAF) and the receptor-independent thapsigargin. Ca(2+)-influx-related increases in [Ca(2+)](i) were calculated by comparing aliquots of fluo-3-loaded neutrophils in the presence and absence of extracellular Ca(2+). Moreover, Mn(2+) quenching was used in fura-2-loaded cells. We compared 8Br-ADPR with ACA. 8Br-ADPR was exclusively effective when Ca(2+) influx (or Mn(2+) quenching) was induced by fMLP; it did not affect influx when PAF or thapsigargin was the stimulus. ACA inhibited Ca(2+) influx significantly more strongly when this was induced by PAF than by fMLP. Moreover, it reduced thapsigargin-induced Ca(2+) influx. The contribution of TRPM2 to Ca(2+) influx in neutrophils strongly depends on the stimulus; it is sizeable in the case of fMLP and minimal in the case of PAF. PAF induces Ca(2+) entry pathways different from TRPM2; the inhibition by ACA suggests the contribution of channels of the TRPC family.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Calcium/metabolism , Cinnamates/pharmacology , TRPM Cation Channels/antagonists & inhibitors , ortho-Aminobenzoates/pharmacology , Adenosine Diphosphate Ribose/pharmacology , Aniline Compounds/chemistry , Fluorescent Dyes/chemistry , Humans , Manganese/chemistry , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Platelet Activating Factor/pharmacology , TRPM Cation Channels/metabolism , Thapsigargin/pharmacology , Xanthenes/chemistry
19.
Anal Biochem ; 393(2): 248-54, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19560435

ABSTRACT

Arginine (Arg)-specific ADP-ribosylation is one of the posttranslational modifications of proteins and is thought to play an important role in reversibly regulating functions of the target proteins in eukaryotes. However, the physiological target protein has not been established. We examined the fragmentation pattern of both ADP-ribosyl-Arg (ADP-R-Arg) and Arg-ADP-ribosylated peptides by quadrupole tandem mass spectrometry and found a specific cleavage of ADP-R-Arg into N-(ADP-ribosyl)-carbodiimide (ADP-R-carbodiimide) and ornithine. Based on this specific fragmentation pattern, we successfully identified the modification site and sequence of Arg-ADP-ribosylated peptide using a two-step collision and showed that ADP-R-carbodiimide is an excellent marker ion for precursor ion scanning of Arg-ADP-ribosylated peptide. We propose that a combination of the precursor ion scanning with ADP-R-carbodiimide as a marker ion and two-step collision is useful in searching for physiological target proteins of Arg-ADP-ribosylation.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Analytic Sample Preparation Methods/methods , Peptides/chemistry , Proteins/chemistry , Sequence Analysis, Protein/methods , Tandem Mass Spectrometry/methods , Adenosine Diphosphate Ribose/analysis , Ornithine/analysis , Poly Adenosine Diphosphate Ribose/chemistry , Protein Processing, Post-Translational
20.
J Am Chem Soc ; 131(20): 6989-96, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19413317

ABSTRACT

A new type of small-molecular sirtuin inhibitor was designed on the basis of the proposed catalytic mechanism for deacetylation of acetylated lysine substrates by sirtuins. Among the compounds thus designed and synthesized, we found that 2k, which contains an ethoxycarbonyl group at the alpha position to the acetamide of acetylated lysine substrate analogue 1, showed potent inhibitory activity in an in vitro assay using recombinant SIRT1, with high selectivity over SIRT2 and SIRT3. Mechanistic study by means of kinetic analysis, mass spectroscopy, and computation indicated that the enol form of compound 2k nucleophilically attacks NAD(+) in the active site of SIRTs to afford the stable compound 2k-ADP-ribose conjugate 5, leading to inhibition of the enzyme activity. Compound 2k also caused a dose-dependent increase of p53 acetylation in human colon cancer HCT116 cells, indicating inhibition of SIRT1 in the cells. These results have implications for the development of selective sirtuin inhibitors by means of mechanism-based drug design.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Lysine/analogs & derivatives , Sirtuins/antagonists & inhibitors , Acetylation , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/pharmacology , HCT116 Cells , Humans , Kinetics , Lysine/chemistry , Lysine/pharmacology , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Sirtuin 1 , Sirtuins/chemistry , Sirtuins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...