Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell Immunol ; 380: 104589, 2022 10.
Article in English | MEDLINE | ID: mdl-36084401

ABSTRACT

The pulp of human teeth contains a population of self-renewing stem cells that can regulate the functions of immune cells. When applied to patients, these cells can protect tissues from damage by excessive inflammation. We confirm that dental pulp cells effectively inhibit the proliferation and activation of cytotoxic T cells in vitro, and show that they carry high levels of CD73, a key enzyme in the conversion of pro-inflammatory extracellular ATP to immunosuppressive adenosine. Given their accessibility and abundance, as well as their potential for allogeneic administration, dental pulp cells provide a valuable source for immunomodulatory therapy.


Subject(s)
Adenosine , Dental Pulp , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Adenosine Triphosphate/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Humans
2.
Int J Mol Sci ; 23(6)2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35328585

ABSTRACT

cADPR is a second messenger that releases Ca2+ from intracellular stores via the ryanodine receptor. Over more than 15 years, it has been controversially discussed whether cADPR also contributes to the activation of the nucleotide-gated cation channel TRPM2. While some groups have observed activation of TRPM2 by cADPR alone or in synergy with ADPR, sometimes only at 37 °C, others have argued that this is due to the contamination of cADPR by ADPR. The identification of a novel nucleotide-binding site in the N-terminus of TRPM2 that binds ADPR in a horseshoe-like conformation resembling cADPR as well as the cADPR antagonist 8-Br-cADPR, and another report that demonstrates activation of TRPM2 by binding of cADPR to the NUDT9H domain raised the question again and led us to revisit the topic. Here we show that (i) the N-terminal MHR1/2 domain and the C-terminal NUDT9H domain are required for activation of human TRPM2 by ADPR and 2'-deoxy-ADPR (2dADPR), (ii) that pure cADPR does not activate TRPM2 under a variety of conditions that have previously been shown to result in channel activation, (iii) the cADPR antagonist 8-Br-cADPR also inhibits activation of TRPM2 by ADPR, and (iv) cADPR does not bind to the MHR1/2 domain of TRPM2 while ADPR does.


Subject(s)
Cyclic ADP-Ribose , TRPM Cation Channels , Binding Sites , Calcium/metabolism , Calcium Signaling , Cyclic ADP-Ribose/metabolism , Humans , TRPM Cation Channels/metabolism
3.
Biochem Soc Trans ; 47(1): 329-337, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30674608

ABSTRACT

Adenine nucleotides (AdNs) play important roles in immunity and inflammation. Extracellular AdNs, such as adenosine triphosphate (ATP) or nicotinamide adenine dinucleotide (NAD) and their metabolites, act as paracrine messengers by fine-tuning both pro- and anti-inflammatory processes. Moreover, intracellular AdNs derived from ATP or NAD play important roles in many cells of the immune system, including T lymphocytes, macrophages, neutrophils and others. These intracellular AdNs are signaling molecules that transduce incoming signals into meaningful cellular responses, e.g. activation of immune responses against pathogens.


Subject(s)
Adenine Nucleotides/metabolism , Inflammation/metabolism , Macrophages/immunology , Neutrophils/immunology , Second Messenger Systems , T-Lymphocytes/immunology , Adenosine Triphosphate/metabolism , Humans , Macrophages/metabolism , NAD/metabolism , Neutrophils/metabolism , Paracrine Communication , Signal Transduction , T-Lymphocytes/metabolism
4.
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
5.
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
6.
Purinergic Signal ; 15(2): 155-166, 2019 06.
Article in English | MEDLINE | ID: mdl-31016551

ABSTRACT

ATP and its metabolites are important extracellular signal transmitters acting on purinergic P2 and P1 receptors. Most cells can actively secrete ATP in response to a variety of external stimuli such as gating of the P2X7 receptor. We used Yac-1 murine lymphoma cells to study P2X7-mediated ATP release. These cells co-express P2X7 and ADP-ribosyltransferase ARTC2, permitting gating of P2X7 by NAD+-dependent ADP-ribosylation without the need to add exogenous ATP. Yac-1 cells released ATP into the extracellular space within minutes after stimulation with NAD+. This was blocked by pre-incubation with the inhibitory P2X7-specific nanobody 13A7. Gating of P2X7 for 3 h significantly decreased intracellular ATP levels in living cells, but these had returned to normal by 20 h. P2X7-mediated ATP release was dependent on a rise in cytosolic calcium and the depletion of intracellular potassium, but was not blocked by inhibitors of pannexins or connexins. We used genetically encoded FRET-based ATP sensors targeted to the cytosol to image P2X7-mediated changes in the distribution of ATP in 3T3 fibroblasts co-expressing P2X7 and ARTC2 and in Yac-1 cells. In response to NAD+, we observed a marked depletion of ATP in the cytosol. This study demonstrates the potential of ATP sensors as tools to study regulated ATP release by other cell types under other conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Receptors, Purinergic P2X7/metabolism , 3T3 Cells , Animals , Cell Line, Tumor , Cytosol/metabolism , Fluorescence Resonance Energy Transfer/methods , Mice
7.
Biochim Biophys Acta Mol Cell Res ; 1864(6): 977-986, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27913206

ABSTRACT

Ca2+ signaling is a major signal transduction pathway involved in T cell activation, but also in apoptosis of T cells. Since T cells make use of several Ca2+-mobilizing second messengers, such as nicotinic acid adenine dinucleotide phosphate, d-myo-inositol 1,4,5-trisphosphate, and cyclic ADP-ribose, we intended to analyze luminal Ca2+ concentration upon cell activation. Mag-Fluo4/AM, a low-affinity Ca2+ dye known to localize to the endoplasmic reticular lumen in many cell types, showed superior brightness and bleaching stability, but, surprisingly, co-localized with mito-tracker, but not with ER-tracker in Jurkat T cells. Thus, we used Mag-Fluo4/AM to monitor the free luminal mitochondrial Ca2+ concentration ([Ca2+]mito) in these cells. Simultaneous analysis of the free cytosolic Ca2+ concentration ([Ca2+]i) and [Ca2+]mito upon cell stimulation revealed that Ca2+ signals in the majority of mitochondria were initiated at [Ca2+ ]i≥approx. 400 to 550nM. In primary murine CD4+ T cells, Mag-Fluo4 showed two different localization patterns: either co-localization with mito-tracker, as in Jurkat T cells, or with ER-tracker. Thus, in single primary murine CD4+ T cells, either decreases of [Ca2+ ]ER or increases of [Ca2+ ]mito were observed upon cell stimulation. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.


Subject(s)
Aniline Compounds/metabolism , Calcium/metabolism , Organelles/metabolism , Xanthenes/metabolism , Humans , Jurkat Cells , Mitochondria/metabolism
8.
Biochem J ; 474(13): 2159-2175, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28515263

ABSTRACT

TRPM2 (transient receptor potential channel, subfamily melastatin, member 2) is a Ca2+-permeable non-selective cation channel activated by the binding of adenosine 5'-diphosphoribose (ADPR) to its cytoplasmic NUDT9H domain (NUDT9 homology domain). Activation of TRPM2 by ADPR downstream of oxidative stress has been implicated in the pathogenesis of many human diseases, rendering TRPM2 an attractive novel target for pharmacological intervention. However, the structural basis underlying this activation is largely unknown. Since ADP (adenosine 5'-diphosphate) alone did not activate or antagonize the channel, we used a chemical biology approach employing synthetic analogues to focus on the role of the ADPR terminal ribose. All novel ADPR derivatives modified in the terminal ribose, including that with the seemingly minor change of methylating the anomeric-OH, abolished agonist activity at TRPM2. Antagonist activity improved as the terminal substituent increasingly resembled the natural ribose, indicating that gating by ADPR might require specific interactions between hydroxyl groups of the terminal ribose and the NUDT9H domain. By mutating amino acid residues of the NUDT9H domain, predicted by modelling and docking to interact with the terminal ribose, we demonstrate that abrogating hydrogen bonding of the amino acids Arg1433 and Tyr1349 interferes with activation of the channel by ADPR. Taken together, using the complementary experimental approaches of chemical modification of the ligand and site-directed mutagenesis of TRPM2, we demonstrate that channel activation critically depends on hydrogen bonding of Arg1433 and Tyr1349 with the terminal ribose. Our findings allow for a more rational design of novel TRPM2 antagonists that may ultimately lead to compounds of therapeutic potential.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Arginine/metabolism , TRPM Cation Channels/metabolism , Tyrosine/metabolism , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/genetics , Amino Acid Sequence , Arginine/chemistry , Arginine/genetics , Calcium/metabolism , Calcium Signaling , HEK293 Cells , Humans , Ion Channel Gating , Mutagenesis, Site-Directed , Mutation/genetics , Patch-Clamp Techniques , Protein Binding , Protein Conformation , Pyrophosphatases/metabolism , Sequence Homology, Amino Acid , TRPM Cation Channels/chemistry , TRPM Cation Channels/genetics , Tyrosine/chemistry , Tyrosine/genetics
9.
J Biol Chem ; 288(22): 16017-30, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23564460

ABSTRACT

Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most potent Ca(2+)-releasing second messenger known to date. Here, we report a new role for NAADP in arrhythmogenic Ca(2+) release in cardiac myocytes evoked by ß-adrenergic stimulation. Infusion of NAADP into intact cardiac myocytes induced global Ca(2+) signals sensitive to inhibitors of both acidic Ca(2+) stores and ryanodine receptors and to NAADP antagonist BZ194. Furthermore, in electrically paced cardiac myocytes BZ194 blocked spontaneous diastolic Ca(2+) transients caused by high concentrations of the ß-adrenergic agonist isoproterenol. Ca(2+) transients were recorded both as increases of the free cytosolic Ca(2+) concentration and as decreases of the sarcoplasmic luminal Ca(2+) concentration. Importantly, NAADP antagonist BZ194 largely ameliorated isoproterenol-induced arrhythmias in awake mice. We provide strong evidence that NAADP-mediated modulation of couplon activity plays a role for triggering spontaneous diastolic Ca(2+) transients in isolated cardiac myocytes and arrhythmias in the intact animal. Thus, NAADP signaling appears an attractive novel target for antiarrhythmic therapy.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Arrhythmias, Cardiac/metabolism , Calcium Signaling/drug effects , Isoproterenol/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , NADP/analogs & derivatives , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/pathology , Cells, Cultured , Mice , Myocardium/pathology , Myocytes, Cardiac/pathology , NADP/antagonists & inhibitors , NADP/metabolism , Nicotinic Acids/pharmacology , Ryanodine Receptor Calcium Release Channel/immunology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology
10.
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
11.
J Biol Chem ; 287(39): 32525-34, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22851169

ABSTRACT

Nicotinic acid adenine dinucleotide phosphate (NAADP) is a ubiquitous second messenger providing a Ca(2+) trigger in a wide range of cell types. However, its metabolism is not well understood. Here, we demonstrate the presence of endogenous NAADP in HeLa cells. CD38, a promiscuous enzyme described to be involved in NAADP metabolism, was not detectable in HeLa cells. In cell-free extracts of HeLa cells, NAADP was degraded to nicotinic acid adenine dinucleotide (NAAD). The enzyme was enriched in membranes (10,000 × g pellet) and displayed characteristics typical of alkaline phosphatase (AP), e.g. pH optimum at 8-9 and sensitivity to the inhibitors L-homoarginine and L-leucine. Importantly, NAADP at physiological concentrations (50-100 nM) was degraded to NAAD. Expression of AP isoenzymes was analyzed in HeLa cells. Based on the results together with inhibitor studies, the placental AP isoform emerged as the best candidate for NAADP degradation in HeLa cells. In contrast to HeLa cells, Jurkat T cells or HEK293 cells did not express any AP isoenzymes and did not display any NAADP 2'-phosphatase activity. Finally, the placental AP isoform was expressed heterologously in HEK293 cells, resulting in reconstitution of NAADP 2'-phosphatase activity in cell-free extracts. On the basis of the results, we provide evidence for AP as the metabolizing enzyme of NAADP in cells that do not express CD38.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Alkaline Phosphatase/biosynthesis , Cell Membrane/enzymology , Gene Expression Regulation, Enzymologic/physiology , Membrane Glycoproteins/metabolism , NADP/analogs & derivatives , ADP-ribosyl Cyclase 1/genetics , Alkaline Phosphatase/genetics , Cell Membrane/genetics , HEK293 Cells , HeLa Cells , Humans , Hydrogen-Ion Concentration , Isoenzymes/biosynthesis , Isoenzymes/genetics , Jurkat Cells , Membrane Glycoproteins/genetics , NADP/genetics , NADP/metabolism
12.
Int J Oncol ; 62(1)2023 01.
Article in English | MEDLINE | ID: mdl-36382671

ABSTRACT

Bitter taste receptors (T2Rs) are G protein­coupled receptors originally detected in the gustatory system. More recently, T2Rs have been shown to be expressed in extra­oral cells eliciting non­gustatory functions. Emerging evidence has suggested a potential role for T2R signaling in diverse pathophysiological conditions, including cancer. -The aim of the present study was to evaluate the expression of T2R14 in pancreatic ductal adenocarcinoma (PDAC) and to assess its involvement in the anticancer effects induced by apigenin, a natural ligand of T2R14. For this purpose, T2R14 expression was explored in PDAC tumor tissue and tumor­derived cell lines. Using the cell lines expressing the highest levels of T2R14, its effects on chemoresponsiveness and migration upon activation with apigenin were investigated in vitro. To the best of our knowledge, the present study was the first to confirm the expression of the T2R family member T2R14 in PDAC. Patients with relatively high levels of T2R14 expression exhibited significantly prolonged overall survival compared with that of patients with low T2R14 expression. Furthermore, novel functions for apigenin were revealed; notably, apigenin was shown to elicit cytotoxic, anti­migratory and chemosensitizing effects to 5­fluoruracil (5­FU) and to 5­FU, leucovorin, irinotecan and oxaliplatin in pancreatic cancer cells. In conclusion, the present study extended the evidence for the anticancer effects of apigenin and strongly indicated the functional relevance of T2R14 in PDAC, even though their respective underlying pathways appear to be independent of each other.


Subject(s)
Apigenin , Taste , Humans , Taste/physiology , Apigenin/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Fluorouracil
13.
J Biol Chem ; 286(6): 4500-10, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21148483

ABSTRACT

Recent studies have shown that inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) possesses important roles in the development of immune cells. IP3KB can be targeted to multiple cellular compartments, among them nuclear localization and binding in close proximity to the plasma membrane. The B isoform is the only IP3K that is almost ubiquitously expressed in mammalian cells. Detailed mechanisms of its targeting regulation will be important in understanding the role of Ins(1,4,5)P(3) phosphorylation on subcellular calcium signaling and compartment-specific initiation of pathways leading to regulatory active higher phosphorylated inositol phosphates. Here, we identified an exportin 1-dependent nuclear export signal ((134)LQRELQNVQV) and characterized the amino acids responsible for nuclear localization of IP3KB ((129)RKLR). These two targeting domains regulate the amount of nuclear IP3KB in cells. We also demonstrated that the localization of IP3KB at the plasma membrane is due to its binding to cortical actin structures. Intriguingly, all three of these targeting activities reside in one small polypeptide segment (amino acids 104-165), which acts as a multitargeting domain (MTD). Finally, a hitherto unknown subnuclear localization of IP3KB could be demonstrated in rapidly growing H1299 cells. IP3KB is specifically enriched at nuclear invaginations extending perpendicular between the apical and basal surface of the nucleus of these flat cells. Such nuclear invaginations are known to be involved in Ins(1,4,5)P(3)-mediated Ca(2+) signaling of the nucleus. Our findings indicate that IP3KB not only regulates cytoplasmic Ca(2+) signals by phosphorylation of subplasmalemmal and cytoplasmic Ins(1,4,5)P(3) but may also be involved in modulating nuclear Ca(2+) signals generated from these nuclear envelope invaginations.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium Signaling/physiology , Cell Membrane/enzymology , Nuclear Envelope/enzymology , Nuclear Export Signals/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Actin Cytoskeleton/genetics , Active Transport, Cell Nucleus/physiology , Calcium/metabolism , Cell Membrane/genetics , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/genetics , Inositol 1,4,5-Trisphosphate/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Nuclear Envelope/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
14.
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
15.
J Biol Chem ; 285(8): 5541-54, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20022963

ABSTRACT

Cellular migration is an essential prerequisite for metastatic dissemination of cancer cells. This study demonstrates that the neuron/testis-specific F-actin-targeted inositol 1,4,5-trisphosphate 3-kinase-A (ITPKA) is ectopically expressed in different human tumor cell lines and during tumor progression in the metastatic tumor model Balb-neuT. High expression of ITPKA increases invasive migration in vitro and metastasis in a xenograft SCID mouse model. Mechanistic studies show that ITPKA promotes migration of tumor cells by two different mechanisms as follows: growth factor independently high levels of ITPKA induce the formation of large cellular protrusions by directly modulating the actin cytoskeleton. The F-actin binding activity of ITPKA stabilizes and bundles actin filaments and thus increases the levels of cellular F-actin. In growth factor-stimulated cells, the catalytically active domain enhances basal ITPKA-induced migration by activating store-operated calcium entry through production of inositol 1,3,4,5-tetrakisphosphate and subsequent inhibition of inositol phosphate 5-phosphatase. These two functional activities of ITPKA stimulating tumor cell migration place the enzyme among the potential targets of anti-metastatic therapy.


Subject(s)
Cell Movement , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Neoplasms, Experimental/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Calcium/metabolism , Cytoskeleton/metabolism , Cytoskeleton/pathology , Hep G2 Cells , Humans , Inositol Phosphates/metabolism , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Transplantation, Heterologous
16.
FEBS J ; 288(23): 6769-6782, 2021 12.
Article in English | MEDLINE | ID: mdl-34189846

ABSTRACT

Nudix hydrolase 9 (NUDT9) is a member of the nucleoside linked to another moiety X (NUDIX) protein superfamily, which hydrolyses a broad spectrum of organic pyrophosphates from metabolic processes. ADP-ribose (ADPR) has been the only known endogenous substrate accepted by NUDT9 so far. The Ca2+ -permeable transient receptor potential melastatin subfamily 2 (TRPM2) channel contains a homologous NUDT9-homology (NUDT9H) domain and is activated by ADPR. Sustained Ca2+ influx via ADPR-activated TRPM2 triggers apoptotic mechanisms. Thus, a precise regulation of cellular ADPR levels by NUDT9 is essential. A detailed characterization of the enzyme-substrate interaction would help to understand the high substrate specificity of NUDT9. Here, we analysed ligand binding to NUDT9 using a variety of biophysical techniques. We identified 2'-deoxy-ADPR as an additional substrate for NUDT9. Similar enzyme kinetics and binding affinities were determined for the two ligands. The high-affinity binding was preserved in NUDT9 containing the mutated NUDIX box derived from the human NUDT9H domain. NMR spectroscopy indicated that ADPR and 2'-deoxy-ADPR bind to the same binding site of NUDT9. Backbone resonance assignment and subsequent molecular docking allowed further characterization of the binding pocket. Substantial conformational changes of NUDT9 upon ligand binding were observed which might allow for the development of NUDT9-based ADPR fluorescence resonance energy transfer sensors that may help with the analysis of ADPR signalling processes in cells in the future.


Subject(s)
Adenosine Diphosphate Ribose/chemistry , Molecular Docking Simulation , Protein Conformation , Pyrophosphatases/chemistry , Adenosine Diphosphate Ribose/metabolism , Binding Sites/genetics , Binding, Competitive , Humans , Kinetics , Ligands , Magnetic Resonance Spectroscopy , Mutation , Protein Binding , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Scattering, Small Angle , Substrate Specificity , X-Ray Diffraction
17.
Front Immunol ; 12: 703574, 2021.
Article in English | MEDLINE | ID: mdl-34539634

ABSTRACT

CD38 is the major NAD+-hydrolyzing ecto-enzyme in most mammals. As a type II transmembrane protein, CD38 is also a promising target for the immunotherapy of multiple myeloma (MM). Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Using phage display technology, we isolated 13 mouse CD38-specific nanobodies from immunized llamas and produced these as recombinant chimeric mouse IgG2a heavy chain antibodies (hcAbs). Sequence analysis assigned these hcAbs to five distinct families that bind to three non-overlapping epitopes of CD38. Members of families 4 and 5 inhibit the GDPR-cyclase activity of CD38. Members of families 2, 4 and 5 effectively induce complement-dependent cytotoxicity against CD38-expressing tumor cell lines, while all families effectively induce antibody dependent cellular cytotoxicity. Our hcAbs present unique tools to assess cytotoxicity mechanisms of CD38-specific hcAbs in vivo against tumor cells and potential off-target effects on normal cells expressing CD38 in syngeneic mouse tumor models, i.e. in a fully immunocompetent background.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Heavy Chains/immunology , Membrane Glycoproteins/immunology , Neoplasms/immunology , ADP-ribosyl Cyclase 1/genetics , Animals , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Humans , Immunoglobulin Heavy Chains/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout
18.
Nat Commun ; 12(1): 5911, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625545

ABSTRACT

Immune cells at sites of inflammation are continuously activated by local antigens and cytokines, and regulatory mechanisms must be enacted to control inflammation. The stepwise hydrolysis of extracellular ATP by ectonucleotidases CD39 and CD73 generates adenosine, a potent immune suppressor. Here we report that human effector CD8 T cells contribute to adenosine production by releasing CD73-containing extracellular vesicles upon activation. These extracellular vesicles have AMPase activity, and the resulting adenosine mediates immune suppression independently of regulatory T cells. In addition, we show that extracellular vesicles isolated from the synovial fluid of patients with juvenile idiopathic arthritis contribute to T cell suppression in a CD73-dependent manner. Our results suggest that the generation of adenosine upon T cell activation is an intrinsic mechanism of human effector T cells that complements regulatory T cell-mediated suppression in the inflamed tissue. Finally, our data underscore the role of immune cell-derived extracellular vesicles in the control of immune responses.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/metabolism , CD8-Positive T-Lymphocytes/metabolism , Extracellular Vesicles/metabolism , GPI-Linked Proteins/metabolism , Immunosuppression Therapy , 5'-Nucleotidase/genetics , Adenosine Triphosphate , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Extracellular Vesicles/immunology , Humans , Inflammation , Lymphocyte Activation , Mice , T-Lymphocytes , T-Lymphocytes, Regulatory/immunology
19.
Sci Signal ; 14(675)2021 03 23.
Article in English | MEDLINE | ID: mdl-33758062

ABSTRACT

NAADP-evoked Ca2+ release through type 1 ryanodine receptors (RYR1) is a major mechanism underlying the earliest signals in T cell activation, which are the formation of Ca2+ microdomains. In our characterization of the molecular machinery underlying NAADP action, we identified an NAADP-binding protein, called hematological and neurological expressed 1-like protein (HN1L) [also known as Jupiter microtubule-associated homolog 2 (JPT2)]. Gene deletion of Hn1l/Jpt2 in human Jurkat and primary rat T cells resulted in decreased numbers of initial Ca2+ microdomains and delayed the onset and decreased the amplitude of global Ca2+ signaling. Photoaffinity labeling demonstrated direct binding of NAADP to recombinant HN1L/JPT2. T cell receptor/CD3-dependent coprecipitation of HN1L/JPT2 with RYRs and colocalization of these proteins suggest that HN1L/JPT2 connects NAADP formation with the activation of RYR channels within the first seconds of T cell activation. Thus, HN1L/JPT2 enables NAADP to activate Ca2+ release from the endoplasmic reticulum through RYR.


Subject(s)
Calcium/metabolism , Membrane Microdomains/metabolism , Microtubule-Associated Proteins/metabolism , NADP/analogs & derivatives , Animals , CD3 Complex/metabolism , Calcium Signaling , Endoplasmic Reticulum/metabolism , Humans , Jurkat Cells , Lymphocyte Activation , Microtubule-Associated Proteins/genetics , NADP/metabolism , Protein Binding , Rats , Receptors, Antigen, T-Cell/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , T-Lymphocytes/metabolism
20.
Biochem J ; 422(1): 139-49, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19492987

ABSTRACT

cADPR (cyclic ADP-ribose) is a universal Ca(2+) mobilizing second messenger. In T-cells cADPR is involved in sustained Ca(2+) release and also in Ca(2+) entry. Potential mechanisms for the latter include either capacitative Ca(2+) entry, secondary to store depletion by cADPR, or direct activation of the non-selective cation channel TRPM2 (transient receptor potential cation channel, subfamily melastatin, member 2). Here we characterize the molecular target of the newly-described membrane-permeant cADPR agonist 8-Br-N(1)-cIDPR (8-bromo-cyclic IDP-ribose). 8-Br-N(1)-cIDPR evoked Ca(2+) signalling in the human T-lymphoma cell line Jurkat and in primary rat T-lymphocytes. Ca(2+) signalling induced by 8-Br-N(1)-cIDPR consisted of Ca(2+) release and Ca(2+) entry. Whereas Ca(2+) release was sensitive to both the RyR (ryanodine receptor) blocker RuRed (Ruthenium Red) and the cADPR antagonist 8-Br-cADPR (8-bromo-cyclic ADP-ribose), Ca(2+) entry was inhibited by the Ca(2+) entry blockers Gd(3+) (gadolinium ion) and SKF-96365, as well as by 8-Br-cADPR. To unravel a potential role for TRPM2 in sustained Ca(2+) entry evoked by 8-Br-N(1)-cIDPR, TRPM2 was overexpressed in HEK (human embryonic kidney)-293 cells. However, though activation by H(2)O(2) was enhanced dramatically in those cells, Ca(2+) signalling induced by 8-Br-N(1)-cIDPR was almost unaffected. Similarly, direct analysis of TRPM2 currents did not reveal activation or co-activation of TRPM2 by 8-Br-N(1)-cIDPR. In summary, the sensitivity to the Ca(2+) entry blockers Gd(3+) and SKF-96365 is in favour of the concept of capacitative Ca(2+) entry, secondary to store depletion by 8-Br-N(1)-cIDPR. Taken together, 8-Br-N(1)-cIDPR appears to be the first cADPR agonist affecting Ca(2+) release and secondary Ca(2+) entry, but without effect on TRPM2.


Subject(s)
Cyclic ADP-Ribose/analogs & derivatives , Inosine Nucleotides/pharmacology , Animals , Calcium Signaling/drug effects , Cell Membrane Permeability/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Gadolinium/pharmacology , Humans , Imidazoles/pharmacology , Inosine Nucleotides/chemical synthesis , Inosine Nucleotides/chemistry , Ion Channel Gating/drug effects , Jurkat Cells , Microinjections , Rats , Ruthenium Red/pharmacology , TRPM Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL