Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
1.
ACS Chem Biol ; 19(5): 1093-1105, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38646883

ABSTRACT

Viral macrodomains that can bind to or hydrolyze protein adenosine diphosphate ribosylation (ADP-ribosylation) have emerged as promising targets for antiviral drug development. Many inhibitor development efforts have been directed against the severe acute respiratory syndrome coronavirus 2 macrodomain 1 (SARS-CoV-2 Mac1). However, potent inhibitors for viral macrodomains are still lacking, with the best inhibitors still in the micromolar range. Based on GS-441524, a remdesivir precursor, and our previous studies, we have designed and synthesized potent binders of SARS-CoV-2 Mac1 and other viral macrodomains including those of Middle East respiratory syndrome coronavirus (MERS-CoV), Venezuelan equine encephalitis virus (VEEV), and Chikungunya virus (CHIKV). We show that the 1'-CN group of GS-441524 promotes binding to all four viral macrodomains tested while capping the 1″-OH of GS-441524-diphosphate-ribose with a simple phenyl ring further contributes to binding. Incorporating these two structural features, the best binders show 20- to 6000-fold increases in binding affinity over ADP-ribose for SARS-CoV-2, MERS-CoV, VEEV, and CHIKV macrodomains. Moreover, building on these potent binders, we have developed two highly sensitive fluorescence polarization tracers that only require nanomolar proteins and can effectively resolve the binding affinities of nanomolar inhibitors. Our findings and probes described here will facilitate future development of more potent viral macrodomain inhibitors.


Subject(s)
Antiviral Agents , Fluorescence Polarization , SARS-CoV-2 , Humans , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/chemistry , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Chikungunya virus/drug effects , COVID-19/virology , COVID-19 Drug Treatment , Encephalitis Virus, Venezuelan Equine/drug effects , Encephalitis Virus, Venezuelan Equine/metabolism , Middle East Respiratory Syndrome Coronavirus , Protein Binding , Protein Domains , SARS-CoV-2/drug effects
2.
Structure ; 32(6): 715-724.e3, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38503292

ABSTRACT

Energy-converting NADH:ubiquinone oxidoreductase, respiratory complex I, is a major enzyme of energy metabolism that couples NADH oxidation and ubiquinone reduction with proton translocation. The NADH oxidation site features different enzymatic activities with various nucleotides. While the kinetics of these reactions are well described, only binding of NAD+ and NADH have been structurally characterized. Here, we report the structures of the electron input module of Aquifex aeolicus complex I with bound ADP-ribose and 3-acetylpyridine adenine dinucleotides at resolutions better than 2.0 Å. ADP-ribose acts as inhibitor by blocking the "ADP-handle" motif essential for nucleotide binding. The pyridine group of APADH is minimally offset from flavin, which could contribute to its poorer suitability as substrate. A comparison with other nucleotide co-structures surprisingly shows that the adenine ribose and the pyrophosphate moiety contribute most to nucleotide binding, thus all adenine dinucleotides share core binding modes to the unique Rossmann-fold in complex I.


Subject(s)
Adenosine Diphosphate Ribose , Electron Transport Complex I , Models, Molecular , Protein Binding , Electron Transport Complex I/metabolism , Electron Transport Complex I/chemistry , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/chemistry , Binding Sites , NAD/metabolism , NAD/chemistry , Crystallography, X-Ray , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Oxidation-Reduction
3.
Front Immunol ; 15: 1294357, 2024.
Article in English | MEDLINE | ID: mdl-38318185

ABSTRACT

TRPM2 is a Ca2+ permeable, non-selective cation channel in the plasma membrane that is involved in the innate immune response regulating, for example, chemotaxis in neutrophils and cytokine secretion in monocytes and macrophages. The intracellular adenine nucleotides ADP-ribose (ADPR) and 2'-deoxy-ADPR (2dADPR) activate the channel, in combination with their co-agonist Ca2+. Interestingly, activation of human TRPM2 (hsTRPM2) by 2dADPR is much more effective than activation by ADPR. However, the underlying mechanism of the nucleotides' differential effect on the channel is not yet fully understood. In this study, we performed whole-cell patch clamp experiments with HEK293 cells heterologously expressing hsTRPM2. We show that 2dADPR has an approx. 4-fold higher Ca2+ sensitivity than ADPR (EC50 = 190 and 690 nM). This allows 2dADPR to activate the channel at lower and thus physiological intracellular Ca2+ concentrations. Kinetic analysis of our data reveals that activation by 2dADPR is faster than activation by ADPR. Mutation in a calmodulin binding N-terminal IQ-like motif in hsTRPM2 completely abrogated channel activation by both agonists. However, mutation of a single amino acid residue (W1355A) in the C-terminus of hsTRPM2, at a site of extensive inter-domain interaction, resulted in slower activation by 2dADPR and neutralized the difference in rate of activation between the two agonists. Taken together, we propose a mechanism by which 2dADPR induces higher hsTRPM2 currents than ADPR by means of faster channel activation. The finding that 2dADPR has a higher Ca2+ sensitivity than ADPR may indicate that 2dADPR rather than ADPR activates hsTRPM2 in physiological contexts such as the innate immune response.


Subject(s)
Adenosine Diphosphate Ribose , TRPM Cation Channels , Humans , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/pharmacology , Calcium Signaling , HEK293 Cells , Kinetics , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
4.
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
5.
Angew Chem Int Ed Engl ; 63(4): e202313317, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-37903139

ABSTRACT

The transfer of an adenosine diphosphate (ADP) ribose moiety to a nucleophilic side chain by consumption of nicotinamide adenine dinucleotide is referred to as ADP-ribosylation, which allows for the spatiotemporal regulation of vital processes such as apoptosis and DNA repair. Recent mass-spectrometry based analyses of the "ADP-ribosylome" have identified histidine as ADP-ribose acceptor site. In order to study this modification, a fully synthetic strategy towards α-configured N(τ)- and N(π)-ADP-ribosylated histidine-containing peptides has been developed. Ribofuranosylated histidine building blocks were obtained via Mukaiyama-type glycosylation and the building blocks were integrated into an ADP-ribosylome derived peptide sequence using fluorenylmethyloxycarbonyl (Fmoc)-based solid-phase peptide synthesis. On-resin installation of the ADP moiety was achieved using phosphoramidite chemistry, and global deprotection provided the desired ADP-ribosylated oligopeptides. The stability under various chemical conditions and resistance against (ADP-ribosyl) hydrolase-mediated degradation has been investigated to reveal that the constructs are stable under various chemical conditions and non-degradable by any of the known ADP-ribosylhydrolases.


Subject(s)
Histidine , Solid-Phase Synthesis Techniques , Histidine/metabolism , Peptides/chemistry , ADP-Ribosylation , Adenosine Diphosphate/metabolism , Adenosine Diphosphate Ribose/chemistry
6.
J Am Chem Soc ; 145(25): 14000-14009, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37315125

ABSTRACT

We report here chemoenzymatic and fully synthetic methodologies to modify aspartate and glutamate side chains with ADP-ribose at specific sites on peptides. Structural analysis of aspartate and glutamate ADP-ribosylated peptides reveals near-quantitative migration of the side chain linkage from the anomeric carbon to the 2″- or 3″-ADP-ribose hydroxyl moieties. We find that this linkage migration pattern is unique to aspartate and glutamate ADP-ribosylation and propose that the observed isomer distribution profile is present in biochemical and cellular environments. After defining distinct stability properties of aspartate and glutamate ADP-ribosylation, we devise methods to install homogenous ADP-ribose chains at specific glutamate sites and assemble glutamate-modified peptides into full-length proteins. By implementing these technologies, we show that histone H2B E2 tri-ADP-ribosylation is able to stimulate the chromatin remodeler ALC1 with similar efficiency to histone serine ADP-ribosylation. Our work reveals fundamental principles of aspartate and glutamate ADP-ribosylation and enables new strategies to interrogate the biochemical consequences of this widespread protein modification.


Subject(s)
Aspartic Acid , Glutamic Acid , Aspartic Acid/metabolism , Glutamic Acid/metabolism , ADP-Ribosylation , Histones/metabolism , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Peptides/chemistry
7.
Org Lett ; 25(21): 3961-3966, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37222442

ABSTRACT

Modifications on the hydroxyl groups of ADP-ribosyl units can provide valuable tools for investigating ADP-ribosylation-related molecular interactions, but the chemical syntheses of these compounds are usually difficult due to their inherent complex structures. In this study, we report a poststage synthetic protocol for accessing novel ADP-2″-deoxyribosyl derivatives through designing a light-induced biomimetic reaction, and SPR assays revealed effective binding of ADP-2″-deoxyribosyl peptides to MacroH2A1.1 with a high affinity (KD = 3.75 × 10-6 M).


Subject(s)
ADP-Ribosylation , Adenosine Diphosphate Ribose , Glycosylation , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Peptides/chemistry , Protein Processing, Post-Translational
8.
Proc Natl Acad Sci U S A ; 120(19): e2215068120, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37126687

ABSTRACT

Poly(ADP-ribose) (PAR) is a homopolymer of adenosine diphosphate ribose that is added to proteins as a posttranslational modification to regulate numerous cellular processes. PAR also serves as a scaffold for protein binding in macromolecular complexes, including biomolecular condensates. It remains unclear how PAR achieves specific molecular recognition. Here, we use single-molecule fluorescence resonance energy transfer (smFRET) to evaluate PAR flexibility under different cation conditions. We demonstrate that, compared to RNA and DNA, PAR has a longer persistence length and undergoes a sharper transition from extended to compact states in physiologically relevant concentrations of various cations (Na+, Mg2+, Ca2+, and spermine4+). We show that the degree of PAR compaction depends on the concentration and valency of cations. Furthermore, the intrinsically disordered protein FUS also served as a macromolecular cation to compact PAR. Taken together, our study reveals the inherent stiffness of PAR molecules, which undergo switch-like compaction in response to cation binding. This study indicates that a cationic environment may drive recognition specificity of PAR.


Subject(s)
Adenosine Diphosphate Ribose , Poly Adenosine Diphosphate Ribose , Poly Adenosine Diphosphate Ribose/chemistry , Poly Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/chemistry , Protein Processing, Post-Translational , Protein Binding , Cell Physiological Phenomena
9.
Methods Mol Biol ; 2609: 111-132, 2023.
Article in English | MEDLINE | ID: mdl-36515833

ABSTRACT

ADP-ribosylation is an ancient modification of proteins, nucleic acids, and other biomolecules found in all kingdoms of life as well as in certain viruses. The regulation of fundamental (patho)physiological processes by ADP-ribosylation, including the cellular stress response, inflammation, and immune response to bacterial and viral pathogens, has created a strong interest into the study of modification establishment and removal to explore novel therapeutic approaches. Beyond ADP-ribosylation in humans, direct targeting of factors that alter host ADP-ribosylation signaling (e.g., viral macrodomains) or utilize ADP-ribosylation to manipulate host cell behavior (e.g., bacterial toxins) were shown to reduce virulence and disease severity. However, the realization of these therapeutic potentials is thus far hampered by the unavailability of simple, high-throughput methods to study the modification "writers" and "erasers" and screen for novel inhibitors.Here, we describe a scalable method for the measurement of (ADP-ribosyl)hydrolase activity. The assay relies on the conversion of ADP-ribose released from a modified substrate by the (ADP-ribosyl)hydrolase under investigation into AMP by the phosphodiesterase NudT5 into bioluminescence via a commercially available detection assay. Moreover, this method can be utilized to study the role of nudix- or ENPP-type phosphodiesterases in ADP-ribosylation processing and may also be adapted to investigate the activity of (ADP-ribosyl)transferases. Overall, this method is applicable for both basic biochemical characterization and screening of large drug libraries; hence, it is highly adaptable to diverse project needs.


Subject(s)
ADP-Ribosylation , Adenosine Diphosphate Ribose , Humans , Adenosine Diphosphate Ribose/chemistry , Proteins/chemistry , Phosphoric Diester Hydrolases/metabolism , Hydrolases/metabolism , Drug Discovery
10.
Methods Mol Biol ; 2609: 75-90, 2023.
Article in English | MEDLINE | ID: mdl-36515830

ABSTRACT

ADP-ribosylation (ADPRylation) is a reversible posttranslational modification resulting in the covalent attachment of ADP-ribose (ADPR) moieties on substrate proteins. Naturally occurring protein motifs and domains, including WWEs, PBZs (PAR binding zinc fingers), and macrodomains, act as "readers" for protein-linked ADPR. Although recombinant, antibody-like ADPR detection reagents containing these readers have facilitated the detection of ADPR, they are limited in their ability to capture the dynamic nature of ADPRylation. Herein, we describe the preparation and use of poly(ADP-ribose) (PAR) Trackers (PAR-Ts)-optimized dimerization-dependent or split-protein reassembly PAR sensors containing a naturally occurring PAR binding domain fused to both halves of dimerization-dependent GFP (ddGFP) or split nano luciferase (NanoLuc), respectively. We also describe how these tools can be used for the detection and quantification of PAR levels in biochemical assays with extracts and in living cells. These protocols will allow users to explore the broad utility of PAR-Ts for detecting PAR in various experimental and biological systems.


Subject(s)
Poly Adenosine Diphosphate Ribose , Ribose , Poly Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/chemistry , ADP-Ribosylation , Protein Processing, Post-Translational , Recombinant Proteins/metabolism
11.
Methods Mol Biol ; 2609: 251-270, 2023.
Article in English | MEDLINE | ID: mdl-36515840

ABSTRACT

ADP-ribosylation is a posttranslational modification (PTM) that has crucial functions in a wide range of cellular processes. Although mass spectrometry (MS) in recent years has emerged as a valuable tool for profiling ADP-ribosylation on a system level, the use of conventional MS methods to profile ADP-ribosylation sites in an unbiased way remains a challenge. Here, we describe a protocol for identification of ADP-ribosylated proteins in vivo on a proteome-wide level, and localization of the amino acid side chains modified with this PTM. The method relies on the enrichment of ADP-ribosylated peptides using the Af1521 macrodomain (Karras GI, Kustatscher G, Buhecha HR, Allen MD, Pugieux C, Sait F, Bycroft M, Ladurner AG, EMBO J 24:1911-1920, 2005), followed by liquid chromatography-high-resolution tandem MS (LC-MS/MS) with electron transfer dissociation-based peptide fragmentation methods, resulting in accurate localization of ADP-ribosylation sites. This protocol explains the step-by-step enrichment and identification of ADP-ribosylated peptides from cell culture to data processing using the MaxQuant software suite.


Subject(s)
Adenosine Diphosphate Ribose , Tandem Mass Spectrometry , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Adenosine Diphosphate Ribose/chemistry , ADP-Ribosylation , Protein Processing, Post-Translational , Proteome/metabolism , Peptides/chemistry
12.
Life Sci Alliance ; 6(1)2023 01.
Article in English | MEDLINE | ID: mdl-36368907

ABSTRACT

The modification of substrates with ADP-ribose (ADPr) is important in, for example, antiviral immunity and cancer. Recently, several reagents were developed to detect ADP-ribosylation; however, it is unknown whether they recognise ADPr, specific amino acid-ADPr linkages, or ADPr with the surrounding protein backbone. We first optimised methods to prepare extracts containing ADPr-proteins and observe that depending on the amino acid modified, the modification is heatlabile. We tested the reactivity of available reagents with diverse ADP-ribosylated protein and RNA substrates and observed that not all reagents are equally suited for all substrates. Next, we determined cross-reactivity with adenylylated RNA, AMPylated proteins, and metabolites, including NADH, which are detected by some reagents. Lastly, we analysed ADP-ribosylation using confocal microscopy, where depending on the fixation method, either mitochondrion, nucleus, or nucleolus is stained. This study allows future work dissecting the function of ADP-ribosylation in cells, both on protein and on RNA substrates, as we optimised sample preparation methods and have defined the reagents suitable for specific methods and substrates.


Subject(s)
ADP-Ribosylation , RNA , RNA/metabolism , Indicators and Reagents , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Proteins/metabolism , Amino Acids/metabolism
13.
J Am Chem Soc ; 144(45): 20582-20589, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36318515

ABSTRACT

We describe the development and optimization of a methodology to prepare peptides and proteins modified on the arginine residue with an adenosine-di-phosphate-ribosyl (ADPr) group. Our method comprises reacting an ornithine containing polypeptide on-resin with an α-linked anomeric isothiourea N-riboside, ensuing installment of a phosphomonoester at the 5'-hydroxyl of the ribosyl moiety followed by the conversion into the adenosine diphosphate. We use this method to obtain four regioisomers of ADP-ribosylated ubiquitin (UbADPr), each modified with an ADP-ribosyl residue on a different arginine position within the ubiquitin (Ub) protein (Arg42, Arg54, Arg72, and Arg74) as the first reported examples of fully synthetic arginine-linked ADPr-modified proteins. We show the chemically prepared Arg-linked UbADPr to be accepted and processed by Legionella enzymes and compare the entire suite of four Arg-linked UbADPr regioisomers in a variety of biochemical experiments, allowing us to profile the activity and selectivity of Legionella pneumophila ligase and hydrolase enzymes.


Subject(s)
Adenosine Diphosphate Ribose , Arginine , Adenosine Diphosphate Ribose/chemistry , Arginine/metabolism , ADP-Ribosylation , Ubiquitin/chemistry , Ubiquitinated Proteins/metabolism , Peptides/chemistry
14.
MAbs ; 14(1): 2095949, 2022.
Article in English | MEDLINE | ID: mdl-35867844

ABSTRACT

Cluster of differentiation 38 (CD38) is an ecto-enzyme expressed primarily on immune cells that metabolize nicotinamide adenine dinucleotide (NAD+) to adenosine diphosphate ribose or cyclic ADP-ribose and nicotinamide. Other substrates of CD38 include nicotinamide adenine dinucleotide phosphate and nicotinamide mononucleotide, a critical NAD+ precursor in the salvage pathway. NAD+ is an important coenzyme involved in several metabolic pathways and is a required cofactor for the function of sirtuins (SIRTs) and poly (adenosine diphosphate-ribose) polymerases. Declines in NAD+ levels are associated with metabolic and inflammatory diseases, aging, and neurodegenerative disorders. To inhibit CD38 enzyme activity and boost NAD+ levels, we developed TNB-738, an anti-CD38 biparatopic antibody that pairs two non-competing heavy chain-only antibodies in a bispecific format. By simultaneously binding two distinct epitopes on CD38, TNB-738 potently inhibited its enzymatic activity, which in turn boosted intracellular NAD+ levels and SIRT activities. Due to its silenced IgG4 Fc, TNB-738 did not deplete CD38-expressing cells, in contrast to the clinically available anti-CD38 antibodies, daratumumab, and isatuximab. TNB-738 offers numerous advantages compared to other NAD-boosting therapeutics, including small molecules, and supplements, due to its long half-life, specificity, safety profile, and activity. Overall, TNB-738 represents a novel treatment with broad therapeutic potential for metabolic and inflammatory diseases associated with NAD+ deficiencies.Abbreviations: 7-AAD: 7-aminoactinomycin D; ADCC: antibody dependent cell-mediated cytotoxicity; ADCP: antibody dependent cell-mediated phagocytosis; ADPR: adenosine diphosphate ribose; APC: allophycocyanin; cADPR: cyclic ADP-ribose; cDNA: complementary DNA; BSA: bovine serum albumin; CD38: cluster of differentiation 38; CDC: complement dependent cytotoxicity; CFA: Freund's complete adjuvant; CHO: Chinese hamster ovary; CCP4: collaborative computational project, number 4; COOT: crystallographic object-oriented toolkit; DAPI: 4',6-diamidino-2-phenylindole; DNA: deoxyribonucleic acid; DSC: differential scanning calorimetry; 3D: three dimensional; εNAD+: nicotinamide 1,N6-ethenoadenine dinucleotide; ECD: extracellular domain; EGF: epidermal growth factor; FACS: fluorescence activated cell sorting; FcγR: Fc gamma receptors; FITC: fluorescein isothiocyanate; HEK: human embryonic kidney; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IgG: immunoglobulin; IFA: incomplete Freund's adjuvant; IFNγ: Interferon gamma; KB: kinetic buffer; kDa: kilodalton; KEGG: kyoto encyclopedia of genes and genomes; LDH: lactate dehydrogenase; M: molar; mM: millimolar; MFI: mean fluorescent intensity; NA: nicotinic acid; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; NAM: nicotinamide; NGS: next-generation sequencing; NHS/EDC: N-Hydroxysuccinimide/ ethyl (dimethylamino propyl) carbodiimide; Ni-NTA: nickel-nitrilotriacetic acid; nL: nanoliter; NK: natural killer; NMN: nicotinamide mononucleotide; OD: optical density; PARP: poly (adenosine diphosphate-ribose) polymerase; PBS: phosphate-buffered saline; PBMC: peripheral blood mononuclear cell; PDB: protein data bank; PE: phycoerythrin; PISA: protein interfaces, surfaces, and assemblies: PK: pharmacokinetics; mol: picomolar; RNA: ribonucleic acid; RLU: relative luminescence units; rpm: rotations per minute; RU: resonance unit; SEC: size exclusion chromatography; SEM: standard error of the mean; SIRT: sirtuins; SPR: surface plasmon resonance; µg: microgram; µM: micromolar; µL: microliter.


Subject(s)
NAD , Sirtuins , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic ADP-Ribose , Humans , Immunoglobulin G , Leukocytes, Mononuclear/metabolism , NAD/chemistry , NAD/metabolism , NADP , Niacinamide , Nicotinamide Mononucleotide , Ribose
15.
J Mol Biol ; 434(16): 167720, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35839840

ABSTRACT

Viral infection in cells triggers a cascade of molecular defense mechanisms to maintain host-cell homoeostasis. One of these mechanisms is ADP-ribosylation, a fundamental post-translational modification (PTM) characterized by the addition of ADP-ribose (ADPr) on substrates. Poly(ADP-ribose) polymerases (PARPs) are implicated in this process and they perform ADP-ribosylation on host and pathogen proteins. Some viral families contain structural motifs that can reverse this PTM. These motifs known as macro domains (MDs) are evolutionarily conserved protein domains found in all kingdoms of life. They are divided in different classes with the viral belonging to Macro-D-type class because of their properties to recognize and revert the ADP-ribosylation. Viral MDs are potential pharmaceutical targets, capable to counteract host immune response. Sequence and structural homology between viral and human MDs are an impediment for the development of new active compounds against their function. Remdesivir, is a drug administrated in viral infections inhibiting viral replication through RNA-dependent RNA polymerase (RdRp). Herein, GS-441524, the active metabolite of the remdesivir, is tested as a hydrolase inhibitor for several viral MDs and for its binding to human homologs found in PARPs. This study presents biochemical and biophysical studies, which indicate that GS-441524 selectively modifies SARS-CoV-2 MD de-MARylation activity, while it does not interact with hPARP14 MD2 and hPARP15 MD2. The structural investigation of MD•GS-441524 complexes, using solution NMR and X-ray crystallography, discloses the impact of certain amino acids in ADPr binding cavity suggesting that F360 and its adjacent residues tune the selective binding of the inhibitor to SARS-CoV-2 MD.


Subject(s)
ADP-Ribosylation , Adenosine/analogs & derivatives , Coronavirus Protease Inhibitors , Poly(ADP-ribose) Polymerases , SARS-CoV-2 , ADP-Ribosylation/drug effects , Adenosine/chemistry , Adenosine/pharmacology , Adenosine Diphosphate Ribose/chemistry , Coronavirus Protease Inhibitors/chemistry , Coronavirus Protease Inhibitors/pharmacology , Humans , Poly(ADP-ribose) Polymerases/chemistry , Protein Binding , Protein Domains , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology
16.
Protein Sci ; 31(6): e4320, 2022 06.
Article in English | MEDLINE | ID: mdl-35634784

ABSTRACT

Transient receptor potential melastatin 2 (TRPM2) is a Ca2+ -permeable, nonselective cation channel involved in diverse physiological processes such as immune response, apoptosis, and body temperature sensing. TRPM2 is activated by ADP-ribose (ADPR) and 2'-deoxy-ADPR in a Ca2+ -dependent manner. While two distinct binding sites exist for ADPR that exert different functions dependent on the species, the involvement of either binding site regarding the superagonistic effect of 2'-deoxy-ADPR is not clear yet. Here, we report the crystal structure of the MHR1/2 domain of TRPM2 from zebrafish (Danio rerio), and show that both ligands bind to this domain and activate the channel. We identified a so far unrecognized Zn2+ -binding domain that was not resolved in previous cryo-EM structures and that is conserved in most TRPM channels. In combination with patch clamp experiments we comprehensively characterize the effect of the Zn2+ -binding domain on TRPM2 activation. Our results provide insight into a conserved motif essential for structural integrity and channel activity.


Subject(s)
TRPM Cation Channels , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Animals , Calcium/metabolism , TRPM Cation Channels/chemistry , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Zebrafish/metabolism , Zinc/metabolism
17.
Methods ; 203: 575-583, 2022 07.
Article in English | MEDLINE | ID: mdl-34560250

ABSTRACT

Protein adenosine diphosphate-ribosylation (ADPr) is caused by the covalent binding of one or more ADP-ribose moieties to a target protein and regulates the biological functions of the target protein. To fully understand the regulatory mechanism of ADP-ribosylation, the essential step is the identification of the ADPr sites from the proteome. As the experimental approaches are costly and time-consuming, it is necessary to develop a computational tool to predict ADPr sites. Recently, serine has been found to be the major residue type for ADP-ribosylation but no predictor is available. In this study, we collected thousands of experimentally validated human ADPr sites on serine residue and constructed several different machine-learning classifiers. We found that the hybrid model, dubbed DeepSADPr, which integrated the one-dimensional convolutional neural network (CNN) with the One-Hot encoding approach and the word-embedding approach, compared favourably to other models in terms of both ten-fold cross-validation and independent test. Its AUC values reached 0.935 for ten-fold cross-validation. Its values of sensitivity, accuracy and Matthews's correlation coefficient reached 0.933, 0.867 and 0.740, respectively, with the fixed specificity value of 0.80. Overall, DeepSADPr is the first classifier for predicting Serine ADPr sites, which is available at http://www.bioinfogo.org/DeepSADPr.


Subject(s)
Protein Processing, Post-Translational , Serine , ADP-Ribosylation , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Humans , Proteome , Serine/metabolism
18.
Acta Pharmacol Sin ; 43(4): 759-770, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34108651

ABSTRACT

Ion channels are ubiquitously expressed in almost all living cells, and are the third-largest category of drug targets, following enzymes and receptors. The transient receptor potential melastatin (TRPM) subfamily of ion channels are important to cell function and survival. Studies have shown upregulation of the TRPM family of ion channels in various brain tumours. Gliomas are the most prevalent form of primary malignant brain tumours with no effective treatment; thus, drug development is eagerly needed. TRPM2 is an essential ion channel for cell function and has important roles in oxidative stress and inflammation. In response to oxidative stress, ADP-ribose (ADPR) is produced, and in turn activates TRPM2 by binding to the NUDT9-H domain on the C-terminal. TRPM2 has been implicated in various cancers and is significantly upregulated in brain tumours. This article reviews the current understanding of TRPM2 in the context of brain tumours and overviews the effects of potential drug therapies targeting TRPM2 including hydrogen peroxide (H2O2), curcumin, docetaxel and selenium, paclitaxel and resveratrol, and botulinum toxin. It is long withstanding knowledge that gliomas are difficult to treat effectively, therefore investigating TRPM2 as a potential therapeutic target for brain tumours may be of considerable interest in the fields of ion channels and pharmacology.


Subject(s)
Brain Neoplasms , TRPM Cation Channels , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Brain Neoplasms/drug therapy , Calcium/metabolism , Humans , Hydrogen Peroxide/pharmacology , Oxidative Stress , TRPM Cation Channels/physiology
19.
Mol Cell ; 81(21): 4552-4567.e8, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34551281

ABSTRACT

ADP-ribose (ADPr) readers are essential components of ADP-ribosylation signaling, which regulates genome maintenance and immunity. The identification and discrimination between monoADPr (MAR) and polyADPr (PAR) readers is difficult because of a lack of suitable affinity-enrichment reagents. We synthesized well-defined ADPr probes and used these for affinity purifications combined with relative and absolute quantitative mass spectrometry to generate proteome-wide MAR and PAR interactomes, including determination of apparent binding affinities. Among the main findings, MAR and PAR readers regulate various common and distinct processes, such as the DNA-damage response, cellular metabolism, RNA trafficking, and transcription. We monitored the dynamics of PAR interactions upon induction of oxidative DNA damage and uncovered the mechanistic connections between ubiquitin signaling and ADP-ribosylation. Taken together, chemical biology enables exploration of MAR and PAR readers using interaction proteomics. Furthermore, the generated MAR and PAR interaction maps significantly expand our current understanding of ADPr signaling.


Subject(s)
ADP-Ribosylation , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate/chemistry , Proteomics/methods , Ubiquitin-Protein Ligases/chemistry , Allosteric Site , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Biotinylation , Cell Communication , DNA Damage , Genetic Techniques , HeLa Cells , Humans , Mass Spectrometry , Mice , Protein Binding , Protein Processing, Post-Translational , Proteome , Signal Transduction , Ubiquitin
20.
Nucleic Acids Res ; 49(18): 10477-10492, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34508355

ABSTRACT

ADP-ribosylation is a modification that targets a variety of macromolecules and regulates a diverse array of important cellular processes. ADP-ribosylation is catalysed by ADP-ribosyltransferases and reversed by ADP-ribosylhydrolases. Recently, an ADP-ribosyltransferase toxin termed 'DarT' from bacteria, which is distantly related to human PARPs, was shown to modify thymidine in single-stranded DNA in a sequence specific manner. The antitoxin of DarT is the macrodomain containing ADP-ribosylhydrolase DarG, which shares striking structural homology with the human ADP-ribosylhydrolase TARG1. Here, we show that TARG1, like DarG, can reverse thymidine-linked DNA ADP-ribosylation. We find that TARG1-deficient human cells are extremely sensitive to DNA ADP-ribosylation. Furthermore, we also demonstrate the first detection of reversible ADP-ribosylation on genomic DNA in vivo from human cells. Collectively, our results elucidate the impact of DNA ADP-ribosylation in human cells and provides a molecular toolkit for future studies into this largely unknown facet of ADP-ribosylation.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , DNA/metabolism , Thiolester Hydrolases/physiology , Adenosine Diphosphate Ribose/chemistry , Bacterial Toxins/metabolism , Cell Line , DNA Repair , DNA Replication , DNA, Single-Stranded/metabolism , Humans , Thiolester Hydrolases/chemistry , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...