Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Science ; 377(6614): eadc8969, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36048923

ABSTRACT

Cyclic adenosine diphosphate (ADP)-ribose (cADPR) isomers are signaling molecules produced by bacterial and plant Toll/interleukin-1 receptor (TIR) domains via nicotinamide adenine dinucleotide (oxidized form) (NAD+) hydrolysis. We show that v-cADPR (2'cADPR) and v2-cADPR (3'cADPR) isomers are cyclized by O-glycosidic bond formation between the ribose moieties in ADPR. Structures of 2'cADPR-producing TIR domains reveal conformational changes that lead to an active assembly that resembles those of Toll-like receptor adaptor TIR domains. Mutagenesis reveals a conserved tryptophan that is essential for cyclization. We show that 3'cADPR is an activator of ThsA effector proteins from the bacterial antiphage defense system termed Thoeris and a suppressor of plant immunity when produced by the effector HopAM1. Collectively, our results reveal the molecular basis of cADPR isomer production and establish 3'cADPR in bacteria as an antiviral and plant immunity-suppressing signaling molecule.


Subject(s)
ADP-ribosyl Cyclase , Adaptor Proteins, Vesicular Transport , Bacteria , Bacterial Proteins , Cyclic ADP-Ribose , Plant Immunity , Toll-Like Receptors , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/genetics , ADP-ribosyl Cyclase/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Bacteria/immunology , Bacteria/virology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cyclic ADP-Ribose/biosynthesis , Cyclic ADP-Ribose/chemistry , Isomerism , NAD/metabolism , Protein Domains , Receptors, Interleukin-1/chemistry , Signal Transduction , Toll-Like Receptors/chemistry , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Tryptophan/chemistry , Tryptophan/genetics
2.
Biomolecules ; 11(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34356669

ABSTRACT

It has recently been demonstrated that the rat poison vacor interferes with mammalian NAD metabolism, because it acts as a nicotinamide analog and is converted by enzymes of the NAD salvage pathway. Thereby, vacor is transformed into the NAD analog vacor adenine dinucleotide (VAD), a molecule that causes cell toxicity. Therefore, vacor may potentially be exploited to kill cancer cells. In this study, we have developed efficient enzymatic and chemical procedures to produce vacor analogs of NAD and nicotinamide riboside (NR). VAD was readily generated by a base-exchange reaction, replacing the nicotinamide moiety of NAD by vacor, catalyzed by Aplysia californica ADP ribosyl cyclase. Additionally, we present the chemical synthesis of the nucleoside version of vacor, vacor riboside (VR). Similar to the physiological NAD precursor, NR, VR was converted to the corresponding mononucleotide (VMN) by nicotinamide riboside kinases (NRKs). This conversion is quantitative and very efficient. Consequently, phosphorylation of VR by NRKs represents a valuable alternative to produce the vacor analog of NMN, compared to its generation from vacor by nicotinamide phosphoribosyltransferase (NamPT).


Subject(s)
Antineoplastic Agents/chemical synthesis , NAD/chemistry , Niacinamide/analogs & derivatives , Phenylurea Compounds/chemistry , Pyridinium Compounds/chemical synthesis , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , Animals , Antineoplastic Agents/pharmacology , Aplysia/enzymology , Cell Proliferation/drug effects , HEK293 Cells , Humans , Niacinamide/chemical synthesis , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism
3.
Cells ; 8(12)2019 12 05.
Article in English | MEDLINE | ID: mdl-31817547

ABSTRACT

: Human CD157/BST-1 and CD38 are dual receptor-enzymes derived by gene duplication that belong to the ADP ribosyl cyclase gene family. First identified over 30 years ago as Mo5 myeloid differentiation antigen and 10 years later as Bone Marrow Stromal Cell Antigen 1 (BST-1), CD157 proved not to be restricted to the myeloid compartment and to have a diversified functional repertoire ranging from immunity to cancer and metabolism. Despite being a NAD+-metabolizing ectoenzyme anchored to the cell surface through a glycosylphosphatidylinositol moiety, the functional significance of human CD157 as an enzyme remains unclear, while its receptor role emerged from its discovery and has been clearly delineated with the identification of its high affinity binding to fibronectin. The aim of this review is to provide an overview of the immunoregulatory functions of human CD157/BST-1 in physiological and pathological conditions. We then focus on CD157 expression in hematological tumors highlighting its emerging role in the interaction between acute myeloid leukemia and extracellular matrix proteins and its potential utility for monoclonal antibody targeted therapy in this disease.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism , ADP-ribosyl Cyclase/antagonists & inhibitors , ADP-ribosyl Cyclase/chemistry , Adaptive Immunity , Antigens, CD/chemistry , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Disease Susceptibility , Enzyme Activation , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , Humans , Immunity, Innate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Models, Molecular , Molecular Targeted Therapy , Myeloid Cells/drug effects , Protein Conformation , Structure-Activity Relationship , Substrate Specificity , Tissue Distribution
4.
Front Immunol ; 10: 2301, 2019.
Article in English | MEDLINE | ID: mdl-31636635

ABSTRACT

Leukemia develops as the result of intrinsic features of the transformed cell, such as gene mutations and derived oncogenic signaling, and extrinsic factors, such as a tumor-friendly, immunosuppressed microenvironment, predominantly in the lymph nodes and the bone marrow. There, high extracellular levels of nucleotides, mainly NAD+ and ATP, are catabolized by different ectonucleotidases, which can be divided in two families according to substrate specificity: on one side those that metabolize NAD+, including CD38, CD157, and CD203a; on the other, those that convert ATP, namely CD39 (and other ENTPDases) and CD73. They generate products that modulate intracellular calcium levels and that activate purinergic receptors. They can also converge on adenosine generation with profound effects, both on leukemic cells, enhancing chemoresistance and homing, and on non-malignant immune cells, polarizing them toward tolerance. This review will first provide an overview of ectonucleotidases expression within the immune system, in physiological and pathological conditions. We will then focus on different hematological malignancies, discussing their role as disease markers and possibly pathogenic agents. Lastly, we will describe current efforts aimed at therapeutic targeting of this family of enzymes.


Subject(s)
Adenosine Triphosphate/metabolism , Hematologic Neoplasms/enzymology , NAD/metabolism , Nucleotidases/physiology , 5'-Nucleotidase/chemistry , 5'-Nucleotidase/physiology , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/physiology , ADP-ribosyl Cyclase 1/chemistry , ADP-ribosyl Cyclase 1/physiology , Animals , Antigens, CD/chemistry , Antigens, CD/physiology , Apyrase/chemistry , Apyrase/physiology , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/physiology , Hematologic Neoplasms/drug therapy , Humans , Nucleotidases/antagonists & inhibitors
5.
Cell Rep ; 27(12): 3684-3695.e4, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31216484

ABSTRACT

cADPR is a well-recognized signaling molecule by modulating the RyRs, but considerable debate exists regarding whether cADPR can bind to and gate the TRPM2 channel, which mediates oxidative stress signaling in diverse physiological and pathological processes. Here, we show that purified cADPR evoked TRPM2 channel currents in both whole-cell and cell-free single-channel recordings and specific binding of cADPR to the purified NUDT9-H domain of TRPM2 by surface plasmon resonance. Furthermore, by combining computational modeling with electrophysiological recordings, we show that the TRPM2 channels carrying point mutations at H1346, T1347, L1379, S1391, E1409, and L1484 possess distinct sensitivity profiles for ADPR and cADPR. These results clearly indicate cADPR is a bona fide activator at the TRPM2 channel and clearly delineate the structural basis for cADPR binding, which not only lead to a better understanding in the gating mechanism of TRPM2 channel but also shed light on a cADPR-induced RyRs-independent Ca2+ signaling mechanism.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Adenosine Diphosphate Ribose/metabolism , Calcium/metabolism , Pyrophosphatases/metabolism , TRPM Cation Channels/metabolism , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/genetics , Binding Sites , HEK293 Cells , Humans , Point Mutation , Protein Conformation , Pyrophosphatases/chemistry , Pyrophosphatases/genetics , TRPM Cation Channels/chemistry , TRPM Cation Channels/genetics
6.
Sci Rep ; 8(1): 15268, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323284

ABSTRACT

Cyclic adenosine 5'-diphosphate ribose (cADPR) is an emerging Ca2+-mobilising second messenger. cADPR analogues have been generated as chemical biology tools via both chemo-enzymatic and total synthetic routes. Both routes rely on the cyclisation of a linear precursor to close an 18-membered macrocyclic ring. We show here that, after cyclisation, there are two possible macrocyclic product conformers that may be formed, depending on whether cyclisation occurs to the "right" or the "left" of the adenine base (as viewed along the H-8 → C-8 base axis). Molecular modelling demonstrates that these two conformers are distinct and cannot interconvert. The two conformers would present a different spatial layout of binding partners to the cADPR receptor/binding site. For chemo-enzymatically generated analogues Aplysia californica ADP-ribosyl cyclase acts as a template to generate solely the "right-handed" conformer and this corresponds to that of the natural messenger, as originally explored using crystallography. However, for a total synthetic analogue it is theoretically possible to generate either product, or a mixture, from a given linear precursor. Cyclisation on either face of the adenine base is broadly illustrated by the first chemical synthesis of the two enantiomers of a "southern" ribose-simplified cIDPR analogue 8-Br-N9-butyl-cIDPR, a cADPR analogue containing only one chiral sugar in the "northern" ribose, i.e. 8-Br-D- and its mirror image 8-Br-L-N9-butyl-cIDPR. By replacing the D-ribose with the unnatural L-ribose sugar, cyclisation of the linear precursor with pyrophosphate closure generates a cyclised product spectroscopically identical, but displaying equal and opposite specific rotation. These findings have implications for cADPR analogue design, synthesis and activity.


Subject(s)
Cyclic ADP-Ribose/analogs & derivatives , Cyclic ADP-Ribose/chemistry , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , Animals , Aplysia/enzymology , Aplysia/metabolism , Crystallography, X-Ray , Cyclic ADP-Ribose/chemical synthesis , Cyclic ADP-Ribose/metabolism , Models, Molecular , Molecular Conformation , Second Messenger Systems , Stereoisomerism
7.
Sci Rep ; 8(1): 1036, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348648

ABSTRACT

Cyclic ADP-ribose (cADPR) is a messenger for Ca2+ mobilization. Its turnover is believed to occur by glycohydrolysis to ADP-ribose. However, ADP-ribose/CDP-alcohol diphosphatase (ADPRibase-Mn) acts as cADPR phosphohydrolase with much lower efficiency than on its major substrates. Recently, we showed that mutagenesis of human ADPRibase-Mn at Phe37, Leu196 and Cys253 alters its specificity: the best substrate of the mutant F37A + L196F + C253A is cADPR by a short difference, Cys253 mutation being essential for cADPR preference. Its proximity to the 'northern' ribose of cADPR in docking models indicates Cys253 is a steric constraint for cADPR positioning. Aiming to obtain a specific cADPR phosphohydrolase, new mutations were tested at Asp250, Val252, Cys253 and Thr279, all near the 'northern' ribose. First, the mutant F37A + L196F + C253G, with a smaller residue 253 (Ala > Gly), showed increased cADPR specificity. Then, the mutant F37A + L196F + V252A + C253G, with another residue made smaller (Val > Ala), displayed the desired specificity, with cADPR kcat/KM ≈20-200-fold larger than for any other substrate. When tested in nucleotide mixtures, cADPR was exhausted while others remained unaltered. We suggest that the specific cADPR phosphohydrolase, by cell or organism transgenesis, or the designed mutations, by genome editing, provide opportunities to study the effect of cADPR depletion on the many systems where it intervenes.


Subject(s)
ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , Cyclic ADP-Ribose/chemistry , Cyclic ADP-Ribose/metabolism , Manganese/chemistry , Manganese/metabolism , ADP-Ribosylation , ADP-ribosyl Cyclase/genetics , Drug Design , Enzyme Activation , Humans , Ligands , Models, Molecular , Mutation , Protein Binding , Structure-Activity Relationship , Substrate Specificity
8.
Sci Rep ; 6: 24213, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27064071

ABSTRACT

ADP-ribosylation is a ubiquitous protein modification utilized by both prokaryotes and eukaryotes for several cellular functions, such as DNA repair, proliferation, and cell signaling. Higher eukaryotes, such as humans, utilize various enzymes to reverse the modification and to regulate ADP-ribose dependent signaling. In contrast, some lower eukaryotes, including trypanosomatids, lack many of these enzymes and therefore have a much more simplified ADP-ribose metabolism. Here we identified and characterized ADP-ribose hydrolases from Trypanosoma brucei and Trypanosoma cruzi, which are homologous to human O-acetyl-ADP-ribose deacetylases MacroD1 and MacroD2. The enzymes are capable for hydrolysis of protein linked ADP-ribose and a product of sirtuin-mediated lysine deacetylation, O-acetyl-ADP-ribose. Crystal structures of the trypanosomatid macrodomains revealed a conserved catalytic site with distinct differences to human MacroD1 and MacroD2.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/enzymology , Trypanosoma cruzi/enzymology , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/genetics , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Binding Sites , Biocatalysis , Calorimetry , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrolases/chemistry , Hydrolases/metabolism , Hydrolysis , Molecular Sequence Data , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment , Sirtuins/metabolism , Thermodynamics
9.
Protein Sci ; 25(3): 650-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26660500

ABSTRACT

Cyclic ADP-ribose (cADPR) mobilizes intracellular Ca(2+) stores and activates Ca(2+) influx to regulate a wide range of physiological processes. It is one of the products produced from the catalysis of NAD(+) by the multifunctional CD38/ADP-ribosyl cyclase superfamily. After elimination of the nicotinamide ring by the enzyme, the reaction intermediate of NAD(+) can either be hydrolyzed to form linear ADPR or cyclized to form cADPR. We have previously shown that human CD38 exhibits a higher preference towards the hydrolysis of NAD(+) to form linear ADPR while Aplysia ADP-ribosyl cyclase prefers cyclizing NAD(+) to form cADPR. In this study, we characterized the enzymatic properties of porcine CD38 and revealed that it has a prominent secondary NAD(+) cyclase activity producing cADPR. We also determined the X-ray crystallographic structures of porcine CD38 and were able to observe conformational flexibility at the base of the active site of the enzyme which allow the NAD(+) reaction intermediate to adopt conformations resulting in both hydrolysis and cyclization forming linear ADPR and cADPR respectively.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , NAD/metabolism , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , ADP-ribosyl Cyclase 1/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Cyclic ADP-Ribose/metabolism , Humans , Models, Molecular , Protein Domains , Swine
10.
J Clin Exp Hematop ; 55(2): 55-60, 2015.
Article in English | MEDLINE | ID: mdl-26490516

ABSTRACT

The identification of eosinophils by flow cytometry is difficult because most of the surface antigens expressed by eosinophils are shared with neutrophils. Some methods have been proposed, generally based on differential light scatter properties, enhanced autofluorescence, lack of CD16 or selective positivity of CD52. Such methods, however, show several limitations. In the present study we report a novel method based on the analysis of glycosylphosphatidylinositol (GPI)-linked molecules. The combination of CD157 and FLAER was used, since FLAER recognizes all GPI-linked molecules, while CD157 is absent on the membrane of eosinophils and expressed by neutrophils. Peripheral blood samples from normal subjects and patients with variable percentages of eosinophils (n = 31), and without any evidence for circulating immature myeloid cells, were stained with the combination of FLAER-Alexa Fluor and CD157-PE. A FascCanto II cytometer was used. Granulocytes were gated after CD33 staining and eosinophils were identified as CD157(-)/FLAER(+) events. Neutrophils were identified as CD157(+)/FLAER(+) events. The percentages of eosinophils detected by this method showed a very significant correlation both with automated counting and with manual counting (r = 0.981 and 0.989, respectively). Sorting assays were carried out by a S3 Cell Sorter: cytospins obtained from CD157(-)/FLAER(+) events consisted of 100% eosinophils, while samples from CD157(+)/FLAER(+) events were represented only by neutrophils. In conclusion, this method shows high sensitivity and specificity in order to distinguish eosinophils from neutrophils by flow cytometry. However, since CD157 is gradually up-regulated throughout bone marrow myeloid maturation, our method cannot be applied to cases characterized by immature myeloid cells.


Subject(s)
ADP-ribosyl Cyclase/chemistry , Antigens, CD/chemistry , Bacterial Toxins/chemistry , Eosinophils/cytology , Flow Cytometry/methods , Pore Forming Cytotoxic Proteins/chemistry , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , CD52 Antigen , Eosinophils/metabolism , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , Glycoproteins/metabolism , Humans , Male , Receptors, IgG/metabolism
11.
Arch Biochem Biophys ; 564: 156-63, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25250980

ABSTRACT

hCD157 catalyzes the hydrolysis of nicotinamide riboside (NR) and nicotinic acid riboside (NAR). The release of nicotinamide or nicotinic acid from NR or NAR was confirmed by spectrophotometric, HPLC and NMR analyses. hCD157 is inactivated by a mechanism-based inhibitor, 2'-deoxy-2'-fluoro-nicotinamide arabinoside (fNR). Modification of the enzyme during the catalytic cycle by NR, NAR, or fNR increased the intrinsic protein fluorescence by approximately 50%. Pre-steady state and steady state data were used to derive a minimal kinetic scheme for the hydrolysis of NR. After initial complex formation a reversible step (360 and 30s(-1)) is followed by a slow irreversible step (0.1s(-1)) that defined the rate limiting step, or kcat. The calculated KMapp value for NR in the hydrolytic reaction is 6nM. The values of the kinetic constants suggest that one biological function of cell-surface hCD157 is to bind and slowly hydrolyze NR, possibly converting it to a ligand-activated receptor. Differences in substrate preference between hCD157 and hCD38 were rationalized through a comparison of the crystal structures of the two proteins. This comparison identified several residues in hCD157 (F108 and F173) that can potentially hinder the binding of dinucleotide substrates (NAD+).


Subject(s)
ADP-ribosyl Cyclase/chemistry , Antigens, CD/chemistry , Niacinamide/analogs & derivatives , Ribonucleosides/chemistry , ADP-ribosyl Cyclase/genetics , ADP-ribosyl Cyclase/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CHO Cells , Catalysis , Cricetinae , Cricetulus , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Hydrolysis , Kinetics , Niacinamide/chemistry , Niacinamide/genetics , Niacinamide/metabolism , Nuclear Magnetic Resonance, Biomolecular , Pyridinium Compounds , Ribonucleosides/genetics , Ribonucleosides/metabolism
12.
J Biol Chem ; 289(22): 15588-601, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24753259

ABSTRACT

CD157/BST-1 behaves both as an ectoenzyme and signaling receptor and is an important regulator of leukocyte trafficking and ovarian cancer progression. However, the molecular interactions underpinning the role of CD157 in these processes remain obscure. The biological functions of CD157 and its partnership with members of the integrin family prompted us to assume the existence of a direct interaction between CD157 and an unknown component of the extracellular matrix. Using solid-phase binding assays and surface plasmon resonance analysis, we demonstrated that CD157 binds fibronectin with high affinity within its heparin-binding domains 1 and 2. Furthermore, we found that CD157 binds to other extracellular matrix proteins containing heparin-binding domains. Finally, we proved that the CD157-fibronectin interaction occurs with living cells, where it elicits CD157-mediated cell responses. Indeed, knockdown of CD157 in Met-5A mesothelial cells changed their morphology and cytoskeleton organization and attenuated the activation of intracellular signaling pathways triggered by fibronectin. This led to impaired cell spreading and adhesion to selected extracellular matrix proteins. Collectively, these findings indicate a central role of CD157 in cell-extracellular matrix interactions and make CD157 an attractive therapeutic target in inflammation and cancer.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Cell Adhesion/physiology , Epithelial Cells/cytology , Fibronectins/metabolism , ADP-ribosyl Cyclase/chemistry , Antigens, CD/chemistry , Cell Differentiation/physiology , Cell Line , Cell Movement/physiology , Epithelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Binding/physiology , Protein Structure, Tertiary , Signal Transduction/physiology , Surface Plasmon Resonance
13.
PLoS One ; 7(4): e34918, 2012.
Article in English | MEDLINE | ID: mdl-22529956

ABSTRACT

Bovine CD38/NAD(+)glycohydrolase (bCD38) catalyses the hydrolysis of NAD(+) into nicotinamide and ADP-ribose and the formation of cyclic ADP-ribose (cADPR). We solved the crystal structures of the mono N-glycosylated forms of the ecto-domain of bCD38 or the catalytic residue mutant Glu218Gln in their apo state or bound to aFNAD or rFNAD, two 2'-fluorinated analogs of NAD(+). Both compounds behave as mechanism-based inhibitors, allowing the trapping of a reaction intermediate covalently linked to Glu218. Compared to the non-covalent (Michaelis) complex, the ligands adopt a more folded conformation in the covalent complexes. Altogether these crystallographic snapshots along the reaction pathway reveal the drastic conformational rearrangements undergone by the ligand during catalysis with the repositioning of its adenine ring from a solvent-exposed position stacked against Trp168 to a more buried position stacked against Trp181. This adenine flipping between conserved tryptophans is a prerequisite for the proper positioning of the N1 of the adenine ring to perform the nucleophilic attack on the C1' of the ribofuranoside ring ultimately yielding cADPR. In all structures, however, the adenine ring adopts the most thermodynamically favorable anti conformation, explaining why cyclization, which requires a syn conformation, remains a rare alternate event in the reactions catalyzed by bCD38 (cADPR represents only 1% of the reaction products). In the Michaelis complex, the substrate is bound in a constrained conformation; the enzyme uses this ground-state destabilization, in addition to a hydrophobic environment and desolvation of the nicotinamide-ribosyl bond, to destabilize the scissile bond leading to the formation of a ribooxocarbenium ion intermediate. The Glu218 side chain stabilizes this reaction intermediate and plays another important role during catalysis by polarizing the 2'-OH of the substrate NAD(+). Based on our structural analysis and data on active site mutants, we propose a detailed analysis of the catalytic mechanism.


Subject(s)
ADP-ribosyl Cyclase 1/chemistry , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Adenosine Monophosphate/chemistry , Amino Acid Substitution , Animals , Catalysis , Catalytic Domain , Cattle , Crystallography, X-Ray , Glycosylation , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , NAD/analogs & derivatives , NAD/chemistry , NAD/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity
14.
Mol Cell Biochem ; 366(1-2): 69-80, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22422046

ABSTRACT

Cyclic ADP-ribose (cADPR), a metabolite of NAD(+), is known to function as a second messenger for intracellular Ca(2+) mobilization in various vertebrate and invertebrate tissues. In this study, we isolated two Xenopus laevis cDNAs (frog cd38 and cd157 cDNAs) homologous to the one encoding the human cADPR-metabolizing enzyme CD38. Frog CD38 and CD157 are 298-amino acid proteins with 35.9 and 27.2 % identity to human CD38 and CD157, respectively. Transfection of expression vectors for frog CD38 and CD157 into COS-7 cells revealed that frog CD38 had NAD(+) glycohydrolase, ADP-ribosyl cyclase (ARC), and cADPR hydrolase activities, and that frog CD157 had no enzymatic activity under physiological conditions. In addition, when recombinant CD38 and frog brain homogenate were electrophoresed on an SDS-polyacrylamide gel, ARC of the brain homogenate migrated to the same position in the gel as that of frog CD38, suggesting that frog CD38 is the major enzyme responsible for cADPR metabolism in amphibian cells. The frog cd38 gene consists of eight exons and is ubiquitously expressed in various tissues. These findings provide evidence for the existence of the CD38-cADPR signaling system in frog cells and suggest that the CD38-cADPR signaling system is conserved during vertebrate evolution.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase/genetics , Antigens, CD/genetics , Cyclic ADP-Ribose/biosynthesis , Xenopus Proteins/genetics , Xenopus laevis/genetics , ADP-ribosyl Cyclase/biosynthesis , ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase 1/biosynthesis , ADP-ribosyl Cyclase 1/chemistry , Amino Acid Sequence , Animals , Antigens, CD/biosynthesis , Antigens, CD/chemistry , Base Sequence , Brain/enzymology , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Conserved Sequence , Cyclic ADP-Ribose/metabolism , Evolution, Molecular , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Humans , Hydrolysis , Inosine Nucleotides/chemistry , Kinetics , Molecular Sequence Data , NAD/analogs & derivatives , NAD/chemistry , Organ Specificity , Phylogeny , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Analysis, DNA , Xenopus Proteins/biosynthesis , Xenopus Proteins/chemistry
15.
J Med Chem ; 55(4): 1478-89, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22248391

ABSTRACT

Two nicotinamide adenine dinucleotide (NAD(+)) analogues modified at the 6 position of the purine ring were synthesized, and their substrate properties toward Aplysia californica ADP-ribosyl cyclase were investigated. 6-N-Methyl NAD(+) (6-N-methyl nicotinamide adenosine 5'-dinucleotide 10) hydrolyzes to give the linear 6-N-methyl ADPR (adenosine 5'-diphosphoribose, 11), whereas 6-thio NHD(+) (nicotinamide 6-mercaptopurine 5'-dinucleotide, 17) generates a cyclic dinucleotide. Surprisingly, NMR correlation spectra confirm this compound to be the N1 cyclic product 6-thio N1-cIDPR (6-thio cyclic inosine 5'-diphosphoribose, 3), although the corresponding 6-oxo analogue is well-known to cyclize at N7. In Jurkat T cells, unlike the parent cyclic inosine 5'-diphosphoribose N1-cIDPR 2, 6-thio N1-cIDPR antagonizes both cADPR- and N1-cIDPR-induced Ca(2+) release but possesses weak agonist activity at higher concentration. 3 is thus identified as the first C-6 modified cADPR (cyclic adenosine 5'-diphosphoribose) analogue antagonist; it represents the first example of a fluorescent N1-cyclized cADPR analogue and is a new pharmacological tool for intervention in the cADPR pathway of cellular signaling.


Subject(s)
Cyclic ADP-Ribose/analogs & derivatives , Cyclic ADP-Ribose/chemical synthesis , T-Lymphocytes/drug effects , Thioinosine/analogs & derivatives , ADP-ribosyl Cyclase/chemistry , Animals , Aplysia , Calcium/metabolism , Cyclic ADP-Ribose/pharmacology , Cyclization , Humans , Hydrogen-Ion Concentration , Jurkat Cells , Models, Molecular , Molecular Conformation , Permeability , Structure-Activity Relationship , T-Lymphocytes/metabolism , Thioinosine/chemical synthesis , Thioinosine/pharmacology
16.
J Mol Biol ; 415(3): 514-26, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22138343

ABSTRACT

Cyclic ADP-ribose (cADPR) is a calcium messenger that can mobilize intracellular Ca²âº stores and activate Ca²âº influx to regulate a wide range of physiological processes. Aplysia cyclase is the first member of the ADP-ribosyl cyclases identified to catalyze the cyclization of NAD⁺ into cADPR. The catalysis involves a two-step reaction, the elimination of the nicotinamide ring and the cyclization of the intermediate resulting in the covalent attachment of the purine ring to the terminal ribose. Aplysia cyclase exhibits a high degree of leniency towards the purine base of its substrate, and the cyclization reaction takes place at either the N1- or the N7-position of the purine ring. To decipher the mechanism of cyclization in Aplysia cyclase, we used a crystallization setup with multiple Aplysia cyclase molecules present in the asymmetric unit. With the use of natural substrates and analogs, not only were we able to capture multiple snapshots during enzyme catalysis resulting in either N1 or N7 linkage of the purine ring to the terminal ribose, we were also able to observe, for the first time, the cyclized products of both N1 and N7 cyclization bound in the active site of Aplysia cyclase.


Subject(s)
ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , Adenosine Diphosphate Ribose/metabolism , Aplysia/enzymology , Animals , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , NAD/metabolism
17.
J Med Chem ; 53(21): 7599-612, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20942470

ABSTRACT

Analogues of nicotinic acid adenine dinucleotide phosphate (NAADP) with substitution at either the 4- or the 5-position position of the nicotinic acid moiety have been synthesized from NADP enzymatically using Aplysia californica ADP-ribosyl cyclase or mammalian NAD glycohydrolase. Substitution at the 4-position of the nicotinic acid resulted in the loss of agonist potency for release of Ca(2+)-ions from sea urchin egg homogenates and in potency for competition ligand binding assays using [(32)P]NAADP. In contrast, several 5-substituted NAADP derivatives showed high potency for binding and full agonist activity for Ca(2+) release. 5-Azido-NAADP was shown to release calcium from sea urchin egg homogenates at low concentration and to compete with [(32)P]NAADP in a competition ligand binding assay with an IC(50) of 18 nM, indicating that this compound might be a potential photoprobe useful for specific labeling and identification of the NAADP receptor.


Subject(s)
Calcium/metabolism , NADP/analogs & derivatives , Niacin/analogs & derivatives , Niacin/chemical synthesis , ADP-ribosyl Cyclase/chemistry , Animals , Aplysia/enzymology , Binding, Competitive , Calcium Channel Agonists/chemical synthesis , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , In Vitro Techniques , NAD+ Nucleosidase/chemistry , NADP/chemical synthesis , NADP/pharmacology , Niacin/pharmacology , Radioligand Assay , Sea Urchins , Structure-Activity Relationship
18.
J Biol Chem ; 284(40): 27637-45, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19640846

ABSTRACT

Cyclic ADP-ribose (cADPR) is a universal calcium messenger molecule that regulates many physiological processes. The production and degradation of cADPR are catalyzed by a family of related enzymes, including the ADP-ribosyl cyclase from Aplysia california (ADPRAC) and CD38 from human. Although ADPRC and CD38 share a common evolutionary ancestor, their enzymatic functions toward NAD and cADPR homeostasis have evolved divergently. Thus, ADPRC can only generate cADPR from NAD (cyclase), whereas CD38, in contrast, has multiple activities, i.e. in cADPR production and degradation, as well as NAD hydrolysis (NADase). In this study, we determined a number of ADPRC and CD38 structures bound with various nucleotides. From these complexes, we elucidated the structural features required for the cyclization (cyclase) reaction of ADPRC and the NADase reaction of CD38. Using the structural approach in combination with site-directed mutagenesis, we identified Phe-174 in ADPRC as a critical residue in directing the folding of the substrate during the cyclization reaction. Thus, a point mutation of Phe-174 to glycine can turn ADPRC from a cyclase toward an NADase. The equivalent residue in CD38, Thr-221, is shown to disfavor the cyclizing folding of the substrate, resulting in NADase being the dominant activity. The comprehensive structural comparison of CD38 and APDRC presented in this study thus provides insights into the structural determinants for the functional evolution from a cyclase to a hydrolase.


Subject(s)
ADP-ribosyl Cyclase/chemistry , ADP-ribosyl Cyclase/metabolism , Evolution, Molecular , NAD/metabolism , ADP-ribosyl Cyclase/genetics , ADP-ribosyl Cyclase 1/chemistry , ADP-ribosyl Cyclase 1/metabolism , Animals , Aplysia/enzymology , Binding Sites , Hydrolysis , Inosine Nucleotides/metabolism , Models, Molecular , Mutation , Protein Conformation , Substrate Specificity
19.
J Phys Chem B ; 112(47): 14682-6, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-18956900

ABSTRACT

Proteins are macromolecules with characteristic structures and biological functions. It is extremely challenging to obtain protein microtube structures through self-assembly as proteins are very complex and flexible. Here we present a strategy showing how a specific protein, ADP-ribosyl cyclase, helically self-assembles from monomers into hexagonal nanochains and further to highly ordered crystalline microtubes. The structures of protein nanochains and consequently self-assembled superlattice were determined by X-ray crystallography at 4.5 A resolution and imaged by scanning electron microscopy. The protein initially forms into dimers that have a fixed size of 5.6 nm, and then, helically self-assembles into 35.6 nm long hexagonal nanochains. One such nanochain consists of six dimers (12 monomers) that stack in order by a pseudo P6(1) screw axis. Seven nanochains produce a series of large-scale assemblies, nanorods, forming the building blocks for microrods. A proposed aging process of microrods results in the formation of hollow microstructures. Synthesis and characterization of large scale self-assembled protein microtubes may pave a new pathway, capable of not only understanding the self-assembly dynamics of biological materials, but also directing design and fabrication of multifunctional nanobuilding blocks with particular applications in biomedical engineering.


Subject(s)
ADP-ribosyl Cyclase/chemistry , Crystallography, X-Ray , Microscopy, Electron, Scanning , Protein Conformation
20.
Arch Biochem Biophys ; 479(2): 114-20, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18835239

ABSTRACT

ADP-ribosyl cyclase and NAD+ glycohydrolase (CD38, E.C.3.2.2.5) efficiently catalyze the exchange of the nicotinamidyl moiety of NAD+, nicotinamide adenine dinucleotide phosphate (NADP+) or nicotinamide mononucleotide (NMN+) with an alternative base. 4'-Pyridinyl drugs (amrinone, milrinone, dismerinone and pinacidil) were efficient alternative substrates (k(cat)/K(M)=0.9-10 microM(-1)s(-1)) in the exchange reaction with ADP-ribosyl cyclase. When CD38 was used as a catalyst the k(cat)/K(M) values for the exchange reaction were reduced two or more orders of magnitude (0.015-0.15 microM(-1)s(-1)). The products of this reaction were novel dinucleotides. The values of the equilibrium constants for dinucleotide formation were determined for several drugs. These enzymes also efficiently catalyze the formation of novel mononucleotides in an exchange reaction with NMN+, k(cat)/K(M)=0.05-0.4 microM(-1)s(-1). The k(cat)/K(M) values for the exchange reaction with NMN+ were generally similar (0.04-0.12 microM(-1)s(-1)) with CD38 and ADP-ribosyl cyclase as catalysts. Several novel heterocyclic alternative substrates were identified as 2-isoquinolines, 1,6-naphthyridines and tricyclic bases. The k(cat)/K(M) values for the exchange reaction with these substrates varied over five orders of magnitude and approached the limit of diffusion with 1,6-naphthyridines. The exchange reaction could be used to synthesize novel mononucleotides or to identify novel reversible inhibitors of CD38.


Subject(s)
ADP-ribosyl Cyclase/chemistry , Aplysia/enzymology , NAD+ Nucleosidase/chemistry , Ribonucleotides/chemistry , ADP-ribosyl Cyclase/metabolism , Animals , Catalysis , Enzyme Inhibitors/chemistry , NAD+ Nucleosidase/metabolism , Ribonucleotides/metabolism , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...