50 results on '"Fliegert, R."'
Search Results
2. Structure of SARS-CoV-2 NSP3 macrodomain in the apo form
- Author
-
Sander, S., primary, Tidow, H., additional, Fliegert, R., additional, and Sandmann, M., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Cellular effects and metabolic stability of N1-cyclic inosine diphosphoribose and its derivatives
- Author
-
Kirchberger, T, Wagner, G, Xu, J, Cordiglieri, C, Wang, P, Gasser, A, Fliegert, R, Bruhn, S, Flügel, A, Lund, F E, Zhang, L-h, Potter, B V L, and Guse, A H
- Published
- 2006
- Full Text
- View/download PDF
4. TSST-1 Induces Th1 or Th2 Differentiation in Naïve CD4+ T Cells in a Dose- and APC-Dependent Manner
- Author
-
BRANDT, K., VAN DER BOSCH, J., FLIEGERT, R., and GEHRING, S.
- Published
- 2002
5. Regulation of calcium signalling by adenine-based second messengers
- Author
-
Fliegert, R., primary, Gasser, A., additional, and Guse, A.H., additional
- Published
- 2007
- Full Text
- View/download PDF
6. TSST-1 Induces Th1 or Th2 Differentiation in Naïve CD4+ T Cells in a Dose- and APC-Dependent Manner.
- Author
-
Brandt, K., Van Der Bosch, J., Fliegert, R., and Gehring, S.
- Subjects
TOXIC shock syndrome ,INTERFERONS ,T cells - Abstract
Superantigens are potent activators of the immune system, causing a variety of diseases, ranging from food poisoning to septic shock. Here, we examined the effects of different toxic shock syndrome toxin 1 (TSST-1) concentrations on the activation, proliferation and synthesis of interferon-γ (IFN-γ) and interleukin-4 (IL-4) in purified naïve human CD4
+ T cells in a serum-free in vitro system. TSST-1 given in low doses (1–10 pg/ml) generates a pronounced T helper 2 (Th2)-like cytokine profile, characterized by elevated IL-4-expressing T-cell populations and reduced IFN-γ-producing populations, whereas higher doses (100 pg/ml) induce a Th1-like profile, with increased expression of IFN-γ and reduced expression of IL-4. These patterns were even more pronounced by adding exogenous cytokines like IL-12 and IL-4 and by the type of antigen-presenting cells (APCs). Thus, B cells induced Th2 shifts, whereas monocytes favoured Th1 induction. Moreover, IL-12 in conditions with B cells counteracted their Th2 bias. Interestingly, in purified naïve T-cell cultures, containing a small population of HLA-DR+ T cells, Th1/Th2 differentiation can be induced by TSST-1 too. There, Th-cell polarization is strongly dependent on TSST-1 concentration, indicating that this is a key parameter in regulating the differentiation of T cells. In conclusion, our data show that Th1/Th2 differentiation of TSST-1-stimulated naïve T cells is controlled by the type of APCs, and in APC-depleted cultures, it depends on the presence of HLA-DR+ cells and TSST-1 concentration. [ABSTRACT FROM AUTHOR]- Published
- 2002
- Full Text
- View/download PDF
7. Synthesis and Biological Evaluation of Novel Membrane-Permeant Cyclic ADP-Ribose Mimics: N<SUP>1</SUP>-[(5 -O-Phosphorylethoxy)methyl]-5-O-phosphorylinosine 5,5 -Cyclicpyrophosphate (cIDPRE) and 8-Substituted Derivatives
- Author
-
Gu, X., Yang, Z., Zhang, L., Kunerth, S., Fliegert, R., Weber, K., Guse, A. H., and Zhang, L.
- Abstract
N1-[(5 -O-Phosphorylethoxy)methyl]-5-O-phosphorylinosine 5,5 -cyclicpyrophosphate (cIDPRE
2a ) and the 8-substituted derivatives 8-bromo-, 8-azido-, 8-amino-, and 8-Cl-cIDPRE (2b −e ) were synthesized from N1-[(5 -acetoxyethoxy)methyl]-2,3-O-isopropylideneinosine (5 ) in good yields. The pharmacological activities of cIDPRE and the 8-substituted derivatives (2a −e ) were analyzed in intact and permeabilized human Jurkat T-lymphocytes. The results indicate that cIDPRE permeates the plasma membrane, releases Ca2+ from an intracellular, cADPR-sensitive Ca2+ store, and subsequently initiates Ca2+ release-activated Ca2+ entry. The Ca2+-releasing activity of cIDPRE was confirmed directly in permeabilized cells. Using time-resolved confocal Ca2+ imaging at the single cell level, the development of global Ca2+ signals starting from local small Ca2+ signals evoked by cIDPRE was observed. 8-N3 -cIDPRE2c and 8-NH2 -cIDPRE2d were similarly effective in their agonistic activity as compared to cIDPRE2a , showing almost indistinguishable concentration−response curves for2a ,2c , and2d and very similar kinetics of Ca2+ signaling. In contrast, the halogenated derivatives 8-Br- and 8-Cl-cIDPRE (2b and2e ) did not significantly elevate [Ca2+]i . Therefore, cIDPRE2a , 8-N3 -cIDPRE2c , and 8-NH2 -cIDPRE2d are novel membrane permeant cADPR mimic and may provide important novel tools to study cADPR-mediated Ca2+ signaling in intact cells.- Published
- 2004
8. 2'-deoxy-ADPR activates human TRPM2 faster than ADPR and thereby induces higher currents at physiological Ca 2+ concentrations.
- Author
-
Pick J, Sander S, Etzold S, Rosche A, Tidow H, Guse AH, and Fliegert R
- Subjects
- Humans, Calcium Signaling, HEK293 Cells, Kinetics, Adenosine Diphosphate Ribose chemistry, Adenosine Diphosphate Ribose pharmacology, TRPM Cation Channels genetics, TRPM Cation Channels metabolism
- Abstract
TRPM2 is a Ca
2+ 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., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Pick, Sander, Etzold, Rosche, Tidow, Guse and Fliegert.)- Published
- 2024
- Full Text
- View/download PDF
9. Potential role of the bitter taste receptor T2R14 in the prolonged survival and enhanced chemoresponsiveness induced by apigenin.
- Author
-
Stern L, Boehme LF, Goetz MR, Nitschke C, Giannou A, Zhang T, Güngör C, Reeh M, Izbicki JR, Fliegert R, Hausen A, Giese N, Hackert T, Niv MY, Heinrich S, Gaida MM, and Ghadban T
- Subjects
- Humans, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Fluorouracil, Taste physiology, Apigenin pharmacology
- Abstract
Bitter taste receptors (T2Rs) are G protein‑coupled receptors originally detected in the gustatory system. More recently, T2Rs have been shown to be expressed in extra‑oral cells eliciting non‑gustatory 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 tumor‑derived 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, anti‑migratory and chemosensitizing effects to 5‑fluoruracil (5‑FU) and to 5‑FU, 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.
- Published
- 2023
- Full Text
- View/download PDF
10. Human dental pulp cells modulate CD8 + T cell proliferation and efficiently degrade extracellular ATP to adenosine in vitro.
- Author
-
Ahmadi P, Yan M, Bauche A, Smeets R, Müller CE, Koch-Nolte F, Haag F, Fliegert R, Kluwe L, Schulze Zur Wiesch J, and Hartjen P
- Subjects
- 5'-Nucleotidase metabolism, Adenosine Triphosphate metabolism, CD8-Positive T-Lymphocytes metabolism, Cell Proliferation, Humans, Adenosine metabolism, Dental Pulp
- 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., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
11. The crystal structure of TRPM2 MHR1/2 domain reveals a conserved Zn 2+ -binding domain essential for structural integrity and channel activity.
- Author
-
Sander S, Pick J, Gattkowski E, Fliegert R, and Tidow H
- Subjects
- Adenosine Diphosphate Ribose chemistry, Adenosine Diphosphate Ribose metabolism, Animals, Calcium metabolism, Zebrafish metabolism, Zinc metabolism, TRPM Cation Channels chemistry, TRPM Cation Channels genetics, TRPM Cation Channels metabolism
- Abstract
Transient receptor potential melastatin 2 (TRPM2) is a Ca
2+ -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., (© 2022 The Authors. Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)- Published
- 2022
- Full Text
- View/download PDF
12. cADPR Does Not Activate TRPM2.
- Author
-
Riekehr WM, Sander S, Pick J, Tidow H, Bauche A, Guse AH, and Fliegert R
- Subjects
- Binding Sites, Calcium metabolism, Calcium Signaling, Humans, Cyclic ADP-Ribose metabolism, TRPM Cation Channels metabolism
- Abstract
cADPR is a second messenger that releases Ca
2+ 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.- Published
- 2022
- Full Text
- View/download PDF
13. Analysis of ligand binding and resulting conformational changes in pyrophosphatase NUDT9.
- Author
-
Gattkowski E, Rutherford TJ, Möckl F, Bauche A, Sander S, Fliegert R, and Tidow H
- Subjects
- 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, Adenosine Diphosphate Ribose chemistry, Molecular Docking Simulation, Protein Conformation, Pyrophosphatases chemistry
- 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 Ca
2+ -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., (© 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2021
- Full Text
- View/download PDF
14. CD73-mediated adenosine production by CD8 T cell-derived extracellular vesicles constitutes an intrinsic mechanism of immune suppression.
- Author
-
Schneider E, Winzer R, Rissiek A, Ricklefs I, Meyer-Schwesinger C, Ricklefs FL, Bauche A, Behrends J, Reimer R, Brenna S, Wasielewski H, Lauten M, Rissiek B, Puig B, Cortesi F, Magnus T, Fliegert R, Müller CE, Gagliani N, and Tolosa E
- Subjects
- 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, 5'-Nucleotidase metabolism, Adenosine metabolism, CD8-Positive T-Lymphocytes metabolism, Extracellular Vesicles metabolism, GPI-Linked Proteins metabolism, Immunosuppression Therapy
- 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., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
15. Mouse CD38-Specific Heavy Chain Antibodies Inhibit CD38 GDPR-Cyclase Activity and Mediate Cytotoxicity Against Tumor Cells.
- Author
-
Baum N, Eggers M, Koenigsdorf J, Menzel S, Hambach J, Staehler T, Fliegert R, Kulow F, Adam G, Haag F, Bannas P, and Koch-Nolte F
- Subjects
- 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, 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
- 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., Competing Interests: FH and FKN receive a share of antibody sales via MediGate GmbH, a wholly owned subsidiary of the University Medical Center Hamburg-Eppendorf. PB and FKN are coinventors on a patent application on CD38-specific nanobodies. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Baum, Eggers, Koenigsdorf, Menzel, Hambach, Staehler, Fliegert, Kulow, Adam, Haag, Bannas and Koch-Nolte.)- Published
- 2021
- Full Text
- View/download PDF
16. HN1L/JPT2: A signaling protein that connects NAADP generation to Ca 2+ microdomain formation.
- Author
-
Roggenkamp HG, Khansahib I, Hernandez C LC, Zhang Y, Lodygin D, Krüger A, Gu F, Möckl F, Löhndorf A, Wolters V, Woike D, Rosche A, Bauche A, Schetelig D, Werner R, Schlüter H, Failla AV, Meier C, Fliegert R, Walseth TF, Flügel A, Diercks BP, and Guse AH
- Subjects
- 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, Calcium metabolism, Membrane Microdomains metabolism, Microtubule-Associated Proteins metabolism, NADP analogs & derivatives
- Abstract
NAADP-evoked Ca
2+ 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., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2021
- Full Text
- View/download PDF
17. Daratumumab and Nanobody-Based Heavy Chain Antibodies Inhibit the ADPR Cyclase but not the NAD + Hydrolase Activity of CD38-Expressing Multiple Myeloma Cells.
- Author
-
Baum N, Fliegert R, Bauche A, Hambach J, Menzel S, Haag F, Bannas P, and Koch-Nolte F
- Abstract
The nucleotides ATP and NAD
+ are released from stressed cells as endogenous danger signals. Ecto-enzymes in the tumor microenvironment hydrolyze these inflammatory nucleotides to immunosuppressive adenosine, thereby, hampering anti-tumor immune responses. The NAD+ hydrolase CD38 is expressed at high levels on the cell surface of multiple myeloma (MM) cells. Daratumumab, a CD38-specific monoclonal antibody promotes cytotoxicity against MM cells. With long CDR3 loops, nanobodies and nanobody-based heavy chain antibodies (hcAbs) might bind to cavities on CD38 and thereby inhibit its enzyme activity more potently than conventional antibodies. The goal of our study was to establish assays for monitoring the enzymatic activities of CD38 on the cell surface of tumor cells and to assess the effects of CD38-specific antibodies on these activities. We monitored the enzymatic activity of CD38-expressing MM and other tumor cell lines, using fluorometric and HPLC assays. Our results showed that daratumumab and hcAb MU1067 inhibit the ADPR cyclase but not the NAD+ hydrolase activity of CD38-expressing MM cells. We conclude that neither clinically approved daratumumab nor recently developed nanobody-derived hcAbs provide a second mode of action against MM cells. Thus, there remains a quest for "double action" CD38-inhibitory antibodies.- Published
- 2020
- Full Text
- View/download PDF
18. Does Cyclic ADP-Ribose (cADPR) Activate the Non-selective Cation Channel TRPM2?
- Author
-
Fliegert R, Riekehr WM, and Guse AH
- Subjects
- Animals, Calcium metabolism, Humans, Protein Binding, Signal Transduction, TRPM Cation Channels genetics, Cyclic ADP-Ribose metabolism, TRPM Cation Channels metabolism
- Abstract
TRPM2 is a non-selective, Ca
2+ -permeable cation channel widely expressed in immune cells. It is firmly established that the channel can be activated by intracellular adenosine 5'-diphosphoribose (ADPR). Until recent cryo-EM structures have exhibited an additional nucleotide binding site in the N-terminus of the channel, this activation was thought to occur via binding to a C-terminal domain of the channel that is highly homologous to the ADPR pyrophosphatase NudT9. Over the years it has been controversially discussed whether the Ca2+ mobilizing second messenger cyclic ADP ribose (cADPR) might also directly activate Ca2+ entry via TRPM2. Here we will review the status of this discussion., (Copyright © 2020 Fliegert, Riekehr and Guse.)- Published
- 2020
- Full Text
- View/download PDF
19. Synthesis of phosphonoacetate analogues of the second messenger adenosine 5'-diphosphate ribose (ADPR).
- Author
-
Baszczyňski O, Watt JM, Rozewitz MD, Fliegert R, Guse AH, and Potter BVL
- Abstract
Adenosine 5'-diphosphate ribose (ADPR) is an intracellular signalling molecule generated from nicotinamide adenine dinucleotide (NAD
+ ). Synthetic ADPR analogues can shed light on the mechanism of activation of ADPR targets and their downstream effects. Such chemical biology studies, however, are often challenging due to the negatively charged pyrophosphate, also sensitive to cellular pyrophosphatases, and prior work on an initial ADPR target, the transient receptor potential cation channel TRPM2, showed complete pyrophosphate group replacement to be a step too far in maintaining biological activity. Thus, we designed ADPR analogues with just one of the negatively charged phosphate groups removed, by employing a phosphonoacetate linker. Synthesis of two novel phosphonoacetate ADPR analogues is described via tandem N,N' -dicyclohexylcarbodiimide coupling to phosphonoacetic acid. Neither analogue, however, showed significant agonist or antagonist activity towards TRPM2, underlining the importance of a complete pyrophosphate motif in activation of this particular receptor.- Published
- 2020
- Full Text
- View/download PDF
20. A structural overview of the ion channels of the TRPM family.
- Author
-
Huang Y, Fliegert R, Guse AH, Lü W, and Du J
- Subjects
- Animals, Binding Sites, Humans, Ion Channel Gating, Ligands, Models, Molecular, Protein Domains, TRPM Cation Channels chemistry
- Abstract
The TRPM (transient receptor potential melastatin) family belongs to the superfamily of TRP cation channels. The TRPM subfamily is composed of eight members that are involved in diverse biological functions such as temperature sensing, inflammation, insulin secretion, and redox sensing. Since the first cloning of TRPM1 in 1998, tremendous progress has been made uncovering the function, structure, and pharmacology of this family. Complete structures of TRPM2, TRPM4, and TRPM8, as well as a partial structure of TRPM7, have been determined by cryo-EM, providing insights into their channel assembly, ion permeation, gating mechanisms, and structural pharmacology. Here we summarize the current knowledge about channel structure, emphasizing general features and principles of the structure of TRPM channels discovered since 2017. We also discuss some of the key unresolved issues in the field, including the molecular mechanisms underlying voltage and temperature dependence, as well as the functions of the TRPM channels' C-terminal domains., (Copyright © 2019 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. NAD binding by human CD38 analyzed by Trp189 fluorescence.
- Author
-
Wolters V, Rosche A, Bauche A, Kulow F, Harneit A, Fliegert R, and Guse AH
- Subjects
- ADP-ribosyl Cyclase 1 genetics, ADP-ribosyl Cyclase 1 metabolism, Binding Sites, HEK293 Cells, Humans, Kinetics, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, NAD metabolism, Tryptophan chemistry, Tryptophan genetics, Tryptophan metabolism, ADP-ribosyl Cyclase 1 chemistry, Membrane Glycoproteins chemistry, NAD chemistry
- Abstract
The NAD-glycohydrolase/ADP-ribosyl cyclase CD38 catalyzes the metabolism of nicotinamide adenine dinucleotide (NAD) to the Ca
2+ mobilizing second messengers ADP-ribose (ADPR), 2'-deoxy-ADPR, and cyclic ADP-ribose (cADPR). In the present study, we investigated binding and metabolism of NAD by a soluble fragment of human CD38, sCD38, and its catalytically inactive mutant by monitoring changes in endogenous tryptophan (Trp) fluorescence. Addition of NAD resulted in a concentration-dependent decrease in sCD38 fluorescence that is mainly caused by the Trp residue W189. Amplitude of the fluorescence decrease was fitted as one-site binding curve revealing a dissociation constant for NAD of 29 μM. A comparable dissociation constant was found with the catalytically inactive sCD38 mutant (KD 37 μM NAD) indicating that binding of NAD is not significantly affected by the mutation. The NAD-induced decrease in Trp fluorescence completely recovered in case of sCD38. Kinetics of recovery was slowed down with decreasing temperature and sCD38 concentration and increasing NAD concentration demonstrating that recovery in fluorescence is proportional to the enzymatic activity of sCD38. Accordingly, recovery in fluorescence was not observed with the catalytically inactive mutant. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
22. Novel CaM-binding motif in its NudT9H domain contributes to temperature sensitivity of TRPM2.
- Author
-
Gattkowski E, Johnsen A, Bauche A, Möckl F, Kulow F, Garcia Alai M, Rutherford TJ, Fliegert R, and Tidow H
- Subjects
- Amino Acid Motifs, HEK293 Cells, Humans, Protein Domains, Protein Stability, TRPM Cation Channels genetics, TRPM Cation Channels metabolism, Hot Temperature, TRPM Cation Channels chemistry
- Abstract
TRPM2 is a non-selective, Ca
2+ -permeable cation channel, which plays a role in cell death but also contributes to diverse immune cell functions. In addition, TRPM2 contributes to the control of body temperature and is involved in perception of non-noxious heat and thermotaxis. TRPM2 is regulated by many factors including Ca2+ , ADPR, 2'-deoxy-ADPR, Ca2+ -CaM, and temperature. However, the molecular basis for the temperature sensitivity of TRPM2 as well as the interplay between the regulatory factors is still not understood. Here we identify a novel CaM-binding site in the unique NudT9H domain of TRPM2. Using a multipronged biophysical approach we show that binding of Ca2+ -CaM to this site occurs upon partial unfolding at temperatures >35 °C and prevents further thermal destabilization. In combination with patch-clamp measurements of full-length TRPM2 our results suggest a role of this CaM-binding site in the temperature sensitivity of TRPM2. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
23. P2X7-mediated ATP secretion is accompanied by depletion of cytosolic ATP.
- Author
-
Johnsen B, Kaschubowski KE, Nader S, Schneider E, Nicola JA, Fliegert R, Wolf IMA, Guse AH, Nikolaev VO, Koch-Nolte F, and Haag F
- Subjects
- 3T3 Cells, Animals, Cell Line, Tumor, Cytosol metabolism, Fluorescence Resonance Energy Transfer methods, Mice, Adenosine Triphosphate metabolism, Receptors, Purinergic P2X7 metabolism
- 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.- Published
- 2019
- Full Text
- View/download PDF
24. Synthesis of Terminal Ribose Analogues of Adenosine 5'-Diphosphate Ribose as Probes for the Transient Receptor Potential Cation Channel TRPM2.
- Author
-
Baszczyňski O, Watt JM, Rozewitz MD, Guse AH, Fliegert R, and Potter BVL
- Subjects
- Chemistry Techniques, Synthetic, HEK293 Cells, Humans, Models, Molecular, Molecular Probes chemistry, Protein Conformation, TRPM Cation Channels chemistry, Adenosine Diphosphate Ribose analogs & derivatives, Molecular Probes chemical synthesis, Molecular Probes metabolism, TRPM Cation Channels metabolism
- 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″-β- 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.
- Published
- 2019
- Full Text
- View/download PDF
25. Correction: Culturing and patch clamping of Jurkat T cells and neurons on Al 2 O 3 coated nanowire arrays of altered morphology.
- Author
-
Harberts J, Zierold R, Fendler C, Koitmäe A, Bayat P, Fernandez-Cuesta I, Loers G, Diercks BP, Fliegert R, Guse AH, Ronning C, Otnes G, Borgström M, and Blick RH
- Abstract
[This corrects the article DOI: 10.1039/C8RA05320K.]., (This journal is © The Royal Society of Chemistry.)
- Published
- 2019
- Full Text
- View/download PDF
26. Culturing and patch clamping of Jurkat T cells and neurons on Al 2 O 3 coated nanowire arrays of altered morphology.
- Author
-
Harberts J, Zierold R, Fendler C, Koitmäe A, Bayat P, Fernandez-Cuesta I, Loers G, Diercks BP, Fliegert R, Guse AH, Ronning C, Otnes G, Borgström M, and Blick RH
- Abstract
Nanowire substrates play an increasingly important role for cell cultures as an approach for hybrid bio-semiconductor junctions. We investigate Jurkat T cells and neurons from mice cultured on Al
2 O3 coated ordered and randomly distributed nanowires. Cell viability was examined by life/membrane staining reporting comparable viability on planar and nanowire substrates. Imaging the hybrid interface reveals a wrapping of the cell membrane around the very nanowire tip. Patch clamp recordings show similar electrophysiological responses on each type of nanowires compared to planar control substrates. We demonstrate that the morphological characteristic of the nanowire substrate plays a subordinate role which opens up the arena for a large range of nanowire substrates in a functionalized application such as stimulation or sensing., Competing Interests: There are no conflicts to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2019
- Full Text
- View/download PDF
27. Adenine nucleotides as paracrine mediators and intracellular second messengers in immunity and inflammation.
- Author
-
Fliegert R, Heeren J, Koch-Nolte F, Nikolaev VO, Lohr C, Meier C, and Guse AH
- Subjects
- Adenosine Triphosphate metabolism, Humans, Macrophages metabolism, NAD metabolism, Neutrophils metabolism, Paracrine Communication, Signal Transduction, T-Lymphocytes metabolism, Adenine Nucleotides metabolism, Inflammation metabolism, Macrophages immunology, Neutrophils immunology, Second Messenger Systems, T-Lymphocytes immunology
- 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., (© 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2019
- Full Text
- View/download PDF
28. TRPM2 activation: Paradigm shifted?
- Author
-
Fliegert R, Hölzer HT, and Guse AH
- Subjects
- Animals, Calcium, Calcium Signaling, Ion Channel Gating, Adenosine Diphosphate Ribose, TRPM Cation Channels
- Abstract
Transient receptor potential cation channel, subtype melastatin 2 (TRPM2), is important for several physiological functions, such as immune response or temperature regulation. Recently, the structure of full-length TRPM2 from zebrafish was published (Huang et al., 2018) proposing a new activation mechanism - is it really a paradigm shift or just reflects evolution of the channel?., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
29. Nanobodies effectively modulate the enzymatic activity of CD38 and allow specific imaging of CD38 + tumors in mouse models in vivo.
- Author
-
Fumey W, Koenigsdorf J, Kunick V, Menzel S, Schütze K, Unger M, Schriewer L, Haag F, Adam G, Oberle A, Binder M, Fliegert R, Guse A, Zhao YJ, Cheung Lee H, Malavasi F, Goldbaum F, van Hegelsom R, Stortelers C, Bannas P, and Koch-Nolte F
- Subjects
- ADP-ribosyl Cyclase 1 immunology, Animals, Camelids, New World, Cell Line, Tumor, Cell Surface Display Techniques, Disease Models, Animal, Epitopes immunology, Fluorescent Dyes, Humans, Membrane Glycoproteins immunology, Mice, Mice, Nude, Multiple Myeloma pathology, Xenograft Model Antitumor Assays, ADP-ribosyl Cyclase 1 metabolism, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Membrane Glycoproteins metabolism, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy, Single-Domain Antibodies immunology
- Abstract
The cell surface ecto-enzyme CD38 is a promising target antigen for the treatment of hematological malignancies, as illustrated by the recent approval of daratumumab for the treatment of multiple myeloma. Our aim was to evaluate the potential of CD38-specific nanobodies as novel diagnostics for hematological malignancies. We successfully identified 22 CD38-specific nanobody families using phage display technology from immunized llamas. Crossblockade analyses and in-tandem epitope binning revealed that the nanobodies recognize three different non-overlapping epitopes, with four nanobody families binding complementary to daratumumab. Three nanobody families inhibit the enzymatic activity of CD38 in vitro, while two others were found to act as enhancers. In vivo, fluorochrome-conjugated CD38 nanobodies efficiently reach CD38 expressing tumors in a rodent model within 2 hours after intravenous injection, thereby allowing for convenient same day in vivo tumor imaging. These nanobodies represent highly specific tools for modulating the enzymatic activity of CD38 and for diagnostic monitoring CD38-expressing tumors.
- Published
- 2017
- Full Text
- View/download PDF
30. 2'-Deoxyadenosine 5'-diphosphoribose is an endogenous TRPM2 superagonist.
- Author
-
Fliegert R, Bauche A, Wolf Pérez AM, Watt JM, Rozewitz MD, Winzer R, Janus M, Gu F, Rosche A, Harneit A, Flato M, Moreau C, Kirchberger T, Wolters V, Potter BVL, and Guse AH
- Subjects
- 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, Adenosine Diphosphate Ribose analogs & derivatives, Clusterin agonists
- Abstract
Transient receptor potential melastatin 2 (TRPM2) is a ligand-gated Ca
2+ -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.- Published
- 2017
- Full Text
- View/download PDF
31. Ligand-induced activation of human TRPM2 requires the terminal ribose of ADPR and involves Arg1433 and Tyr1349.
- Author
-
Fliegert R, Watt JM, Schöbel A, Rozewitz MD, Moreau C, Kirchberger T, Thomas MP, Sick W, Araujo AC, Harneit A, Potter BVL, and Guse AH
- Subjects
- 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, Adenosine Diphosphate Ribose metabolism, Arginine metabolism, TRPM Cation Channels metabolism, Tyrosine metabolism
- Abstract
TRPM2 (transient receptor potential channel, subfamily melastatin, member 2) is a Ca
2+ -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., (© 2017 The Author(s).)- Published
- 2017
- Full Text
- View/download PDF
32. Mag-Fluo4 in T cells: Imaging of intra-organelle free Ca 2+ concentrations.
- Author
-
Diercks BP, Fliegert R, and Guse AH
- Subjects
- Humans, Jurkat Cells, Mitochondria metabolism, Aniline Compounds metabolism, Calcium metabolism, Organelles metabolism, Xanthenes metabolism
- Abstract
Ca
2+ 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., (Copyright © 2016 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
33. Frontrunners of T cell activation: Initial, localized Ca2+ signals mediated by NAADP and the type 1 ryanodine receptor.
- Author
-
Wolf IM, Diercks BP, Gattkowski E, Czarniak F, Kempski J, Werner R, Schetelig D, Mittrücker HW, Schumacher V, von Osten M, Lodygin D, Flügel A, Fliegert R, and Guse AH
- Subjects
- Aniline Compounds chemistry, Animals, Benzofurans chemistry, Cells, Cultured, Fluorometry methods, Humans, Imidazoles chemistry, Jurkat Cells, Lymphocyte Activation drug effects, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Fluorescence, Muromonab-CD3 pharmacology, NADP metabolism, Ryanodine Receptor Calcium Release Channel genetics, T-Lymphocytes drug effects, TRPM Cation Channels metabolism, Time Factors, Xanthenes chemistry, Calcium metabolism, Calcium Signaling, NADP analogs & derivatives, Ryanodine Receptor Calcium Release Channel metabolism, T-Lymphocytes metabolism
- Abstract
The activation of T cells is the fundamental on switch for the adaptive immune system. Ca(2+) signaling is essential for T cell activation and starts as initial, short-lived, localized Ca(2+) signals. The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) forms rapidly upon T cell activation and stimulates early Ca(2+) signaling. We developed a high-resolution imaging technique using multiple fluorescent Ca(2+) indicator dyes to characterize these early signaling events and investigate the channels involved in NAADP-dependent Ca(2+) signals. In the first seconds of activation of either primary murine T cells or human Jurkat cells with beads coated with an antibody against CD3, we detected Ca(2+) signals with diameters close to the limit of detection and that were close to the activation site at the plasma membrane. In Jurkat cells in which the ryanodine receptor (RyR) was knocked down or in primary T cells from RyR1(-/-) mice, either these early Ca(2+) signals were not detected or the number of signals was markedly reduced. Local Ca(2+) signals observed within 20 ms upon microinjection of Jurkat cells with NAADP were also sensitive to RyR knockdown. In contrast, TRPM2 (transient receptor potential channel, subtype melastatin 2), a potential NAADP target channel, was not required for the formation of initial Ca(2+) signals in primary T cells. Thus, through our high-resolution imaging method, we characterized early Ca(2+) release events in T cells and obtained evidence for the involvement of RyR and NAADP in such signals., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
34. NAD derived second messengers: Role in spontaneous diastolic Ca(2+) transients in murine cardiac myocytes.
- Author
-
Warszta D, Nebel M, Fliegert R, and Guse AH
- Subjects
- ADP-ribosyl Cyclase antagonists & inhibitors, Adrenergic beta-Agonists pharmacology, Animals, Biochemistry methods, Calcium Signaling, Cells, Cultured, Electric Stimulation, Fluorescent Dyes analysis, Fluorescent Dyes metabolism, Fura-2 analogs & derivatives, Fura-2 analysis, Fura-2 metabolism, Heart Ventricles cytology, Heterocyclic Compounds, 4 or More Rings pharmacology, Indoles analysis, Indoles metabolism, Isoproterenol pharmacology, Mice, Inbred C57BL, Myocytes, Cardiac drug effects, Sarcoplasmic Reticulum metabolism, Calcium analysis, Calcium metabolism, Myocytes, Cardiac metabolism, NAD metabolism, Second Messenger Systems
- Abstract
Strong β-adrenergic stimulation induced spontaneous diastolic Ca(2+) transients (SCTs) in electrically paced murine cardiac myocytes [28]. To obtain further insights into the underlying mechanism, we developed a method for a simultaneous analysis, in which the free luminal Ca(2+) concentration in the sarcoplasmic reticulum (SR) ([Ca(2+)]SR) and the free cytosolic Ca(2+) concentration ([Ca(2+)]i) were measured in parallel in the same cell. Each spontaneous diastolic Ca(2+) transient was exactly mirrored by a decrease of [Ca(2+)]SR. Since antagonism of the Ca(2+) mobilizing second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) was shown to block SCTs in single cardiac myocytes [28], we analyzed the effect of the novel ADP-ribosyl cyclase inhibitor SAN4825 on both cytosolic and intra-luminal Ca(2+) transients upon strong β-adrenergic stimulation. A strong antagonist effect of SAN4825 on SCTs at low micromolar concentrations was observed. Our results suggest that the underlying mechanism of spontaneous diastolic Ca(2+) transients observed upon strong β-adrenergic stimulation is sensitization of type 2 ryanodine receptor by the Ca(2+) releasing activity of the products of ADP-ribosyl cyclase activity., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
35. Structure-activity relationship of adenosine 5'-diphosphoribose at the transient receptor potential melastatin 2 (TRPM2) channel: rational design of antagonists.
- Author
-
Moreau C, Kirchberger T, Swarbrick JM, Bartlett SJ, Fliegert R, Yorgan T, Bauche A, Harneit A, Guse AH, and Potter BV
- Subjects
- Adenosine Diphosphate Ribose chemical synthesis, Adenosine Diphosphate Ribose chemistry, Dose-Response Relationship, Drug, Humans, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Adenosine Diphosphate Ribose pharmacology, Drug Design, TRPM Cation Channels antagonists & inhibitors
- Abstract
Adenosine 5'-diphosphoribose (ADPR) activates TRPM2, a Ca(2+), Na(+), and K(+) permeable cation channel. Activation is induced by ADPR binding to the cytosolic C-terminal NudT9-homology domain. To generate the first structure-activity relationship, systematically modified ADPR analogues were designed, synthesized, and evaluated as antagonists using patch-clamp experiments in HEK293 cells overexpressing human TRPM2. Compounds with a purine C8 substituent show antagonist activity, and an 8-phenyl substitution (8-Ph-ADPR, 5) is very effective. Modification of the terminal ribose results in a weak antagonist, whereas its removal abolishes activity. An antagonist based upon a hybrid structure, 8-phenyl-2'-deoxy-ADPR (86, IC50 = 3 μM), is more potent than 8-Ph-ADPR (5). Initial bioisosteric replacement of the pyrophosphate linkage abolishes activity, but replacement of the pyrophosphate and the terminal ribose by a sulfamate-based group leads to a weak antagonist, a lead to more drug-like analogues. 8-Ph-ADPR (5) inhibits Ca(2+) signalling and chemotaxis in human neutrophils, illustrating the potential for pharmacological intervention at TRPM2.
- Published
- 2013
- Full Text
- View/download PDF
36. A mathematical model of T lymphocyte calcium dynamics derived from single transmembrane protein properties.
- Author
-
Schmeitz C, Hernandez-Vargas EA, Fliegert R, Guse AH, and Meyer-Hermann M
- Abstract
Fate decision processes of T lymphocytes are crucial for health and disease. Whether a T lymphocyte is activated, divides, gets anergic, or initiates apoptosis depends on extracellular triggers and intracellular signaling. Free cytosolic calcium dynamics plays an important role in this context. The relative contributions of store-derived calcium entry and calcium entry from extracellular space to T lymphocyte activation are still a matter of debate. Here we develop a quantitative mathematical model of T lymphocyte calcium dynamics in order to establish a tool which allows to disentangle cause-effect relationships between ion fluxes and observed calcium time courses. The model is based on single transmembrane protein characteristics which have been determined in independent experiments. This reduces the number of unknown parameters in the model to a minimum and ensures the predictive power of the model. Simulation results are subsequently used for an analysis of whole cell calcium dynamics measured under various experimental conditions. The model accounts for a variety of these conditions, which supports the suitability of the modeling approach. The simulation results suggest a model in which calcium dynamics dominantly relies on the opening of channels in calcium stores while calcium entry through calcium-release activated channels (CRAC) is more associated with the maintenance of the T lymphocyte calcium levels and prevents the cell from calcium depletion. Our findings indicate that CRAC guarantees a long-term stable calcium level which is required for cell survival and sustained calcium enhancement.
- Published
- 2013
- Full Text
- View/download PDF
37. Adenine Dinucleotide Second Messengers and T-lymphocyte Calcium Signaling.
- Author
-
Ernst IM, Fliegert R, and Guse AH
- Abstract
Calcium signaling is a universal signal transduction mechanism in animal and plant cells. In mammalian T-lymphocytes calcium signaling is essential for activation and re-activation and thus important for a functional immune response. Since many years it has been known that both calcium release from intracellular stores and calcium entry via plasma membrane calcium channels are involved in shaping spatio-temporal calcium signals. Second messengers derived from the adenine dinucleotides NAD and NADP have been implicated in T cell calcium signaling. Nicotinic acid adenine dinucleotide phosphate (NAADP) acts as a very early second messenger upon T cell receptor/CD3 engagement, while cyclic ADP-ribose (cADPR) is mainly involved in sustained partial depletion of the endoplasmic reticulum by stimulating calcium release via ryanodine receptors. Finally, adenosine diphosphoribose (ADPR) a breakdown product of both NAD and cADPR activates a plasma membrane cation channel termed TRPM2 thereby facilitating calcium (and sodium) entry into T cells. Receptor-mediated formation, metabolism, and mode of action of these novel second messengers in T-lymphocytes will be reviewed.
- Published
- 2013
- Full Text
- View/download PDF
38. Nicotinic acid adenine dinucleotide phosphate (NAADP)-mediated calcium signaling and arrhythmias in the heart evoked by β-adrenergic stimulation.
- Author
-
Nebel M, Schwoerer AP, Warszta D, Siebrands CC, Limbrock AC, Swarbrick JM, Fliegert R, Weber K, Bruhn S, Hohenegger M, Geisler A, Herich L, Schlegel S, Carrier L, Eschenhagen T, Potter BV, Ehmke H, and Guse AH
- Subjects
- 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, Adrenergic beta-Agonists pharmacology, Arrhythmias, Cardiac metabolism, Calcium Signaling drug effects, Isoproterenol pharmacology, Myocardium metabolism, Myocytes, Cardiac metabolism, NADP analogs & derivatives
- 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 β-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 β-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.
- Published
- 2013
- Full Text
- View/download PDF
39. NAADP signaling revisited.
- Author
-
Guse AH, Ernst IM, and Fliegert R
- Subjects
- Animals, Calcium metabolism, Humans, NADP metabolism, NADP analogs & derivatives, Signal Transduction
- Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most potent Ca²⁺ mobilizing endogenous compound known to date. Although its Ca²⁺ releasing activity has been demonstrated in many cell types and in response to different extracellular stimuli, several aspects of NAADP signaling are unclear. This overview focuses on the controversial aspects and reviews NAADP's role as second messenger: endogenous concentrations and its receptor-mediated alterations, metabolism, and potential organelle and ion channel targets. Finally, the role of NAADP as Ca²⁺ trigger is discussed by reviewing the development of local into global Ca²⁺ signals evoked by NAADP.
- Published
- 2013
- Full Text
- View/download PDF
40. Nicotinic acid adenine dinucleotide phosphate (NAADP) degradation by alkaline phosphatase.
- Author
-
Schmid F, Fliegert R, Westphal T, Bauche A, and Guse AH
- Subjects
- 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, ADP-ribosyl Cyclase 1 metabolism, Alkaline Phosphatase biosynthesis, Cell Membrane enzymology, Gene Expression Regulation, Enzymologic physiology, Membrane Glycoproteins metabolism, NADP analogs & derivatives
- 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.
- Published
- 2012
- Full Text
- View/download PDF
41. Alternative splicing of the N-terminal cytosolic and transmembrane domains of P2X7 controls gating of the ion channel by ADP-ribosylation.
- Author
-
Schwarz N, Drouot L, Nicke A, Fliegert R, Boyer O, Guse AH, Haag F, Adriouch S, and Koch-Nolte F
- Subjects
- Adenosine Diphosphate Ribose genetics, Adenosine Triphosphate genetics, Adenosine Triphosphate metabolism, Animals, HEK293 Cells, Humans, Mice, Mice, Inbred BALB C, NAD genetics, NAD metabolism, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Structure, Tertiary, Receptors, Purinergic P2X7 genetics, T-Lymphocytes, Regulatory cytology, Adenosine Diphosphate Ribose metabolism, Alternative Splicing physiology, Ion Channel Gating physiology, Protein Processing, Post-Translational physiology, Receptors, Purinergic P2X7 metabolism, T-Lymphocytes, Regulatory metabolism
- Abstract
P2X7 is a homotrimeric ion channel with two transmembrane domains and a large extracellular ATP-binding domain. It plays a key role in the response of immune cells to danger signals released from cells at sites of inflammation. Gating of murine P2X7 can be induced by the soluble ligand ATP, as well as by NAD(+)-dependent ADP-ribosylation of arginine 125, a posttranslational protein modification catalyzed by the toxin-related ecto-enzymes ART2.1 and ART2.2. R125 is located at the edge of the ligand-binding crevice. Recently, an alternative splice variant of P2X7, designated P2X7(k), was discovered that differs from the previously described variant P2X7(a) in the N-terminal 42 amino acid residues composing the first cytosolic domain and most of the Tm1 domain. Here we compare the two splice variants of murine P2X7 with respect to their sensitivities to gating by ADP-ribosylation in transfected HEK cells. Our results show that the P2X7(k) variant is sensitive to activation by ADP-ribosylation whereas the P2X7(a) variant is insensitive, despite higher cell surface expression levels. Interestingly, a single point mutation (R276K) renders the P2X7(a) variant sensitive to activation by ADP-ribosylation. Residue 276 is located at the interface of neighboring subunits approximately halfway between the ADP-ribosylation site and the transmembrane domains. Moreover, we show that naive and regulatory T cells preferentially express the more sensitive P2X7(k) variant, while macrophages preferentially express the P2X7(a) variant. Our results indicate that differential splicing of alternative exons encoding the N-terminal cytosolic and transmembrane domains of P2X7 control the sensitivity of different immune cells to extracellular NAD(+) and ATP.
- Published
- 2012
- Full Text
- View/download PDF
42. Human inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) is a nucleocytoplasmic shuttling protein specifically enriched at cortical actin filaments and at invaginations of the nuclear envelope.
- Author
-
Nalaskowski MM, Fliegert R, Ernst O, Brehm MA, Fanick W, Windhorst S, Lin H, Giehler S, Hein J, Lin YN, and Mayr GW
- Subjects
- 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, Actin Cytoskeleton metabolism, Calcium Signaling physiology, Cell Membrane enzymology, Nuclear Envelope enzymology, Nuclear Export Signals physiology, Phosphotransferases (Alcohol Group Acceptor) metabolism
- 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.
- Published
- 2011
- Full Text
- View/download PDF
43. Inositol 1,4,5-trisphosphate 3-kinase-A is a new cell motility-promoting protein that increases the metastatic potential of tumor cells by two functional activities.
- Author
-
Windhorst S, Fliegert R, Blechner C, Möllmann K, Hosseini Z, Günther T, Eiben M, Chang L, Lin HY, Fanick W, Schumacher U, Brandt B, and Mayr GW
- Subjects
- 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, Cell Movement, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Neoplasm Proteins metabolism, Neoplasms, Experimental enzymology, Phosphotransferases (Alcohol Group Acceptor) metabolism
- 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.
- Published
- 2010
- Full Text
- View/download PDF
44. 8-Bromo-cyclic inosine diphosphoribose: towards a selective cyclic ADP-ribose agonist.
- Author
-
Kirchberger T, Moreau C, Wagner GK, Fliegert R, Siebrands CC, Nebel M, Schmid F, Harneit A, Odoardi F, Flügel A, Potter BV, and Guse AH
- Subjects
- 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, Cyclic ADP-Ribose analogs & derivatives, Inosine Nucleotides pharmacology
- 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.
- Published
- 2009
- Full Text
- View/download PDF
45. Activation of the P2X7 ion channel by soluble and covalently bound ligands.
- Author
-
Schwarz N, Fliegert R, Adriouch S, Seman M, Guse AH, Haag F, and Koch-Nolte F
- Abstract
The homotrimeric P2X7 purinergic receptor has sparked interest because of its capacity to sense adenosine triphosphate (ATP) and nicotinamide adenine dinucleotide (NAD) released from cells and to induce calcium signaling and cell death. Here, we examine the response of arginine mutants of P2X7 to soluble and covalently bound ligands. High concentrations of ecto-ATP gate P2X7 by acting as a soluble ligand and low concentrations of ecto-NAD gate P2X7 following ADP-ribosylation at R125 catalyzed by toxin-related ecto-ADP-ribosyltransferase ART2.2. R125 lies on a prominent cysteine-rich finger at the interface of adjacent receptor subunits, and ADP-ribosylation at this site likely places the common adenine nucleotide moiety into the ligand-binding pocket of P2X7.
- Published
- 2009
- Full Text
- View/download PDF
46. ADP-ribosylation at R125 gates the P2X7 ion channel by presenting a covalent ligand to its nucleotide binding site.
- Author
-
Adriouch S, Bannas P, Schwarz N, Fliegert R, Guse AH, Seman M, Haag F, and Koch-Nolte F
- Subjects
- Amino Acid Substitution, Arginine metabolism, Cell Line, Tumor, Humans, Leukocytes enzymology, Ligands, Models, Molecular, Protein Processing, Post-Translational, Receptors, Purinergic P2X7, ADP Ribose Transferases physiology, Binding Sites physiology, Ion Channel Gating physiology, Nucleotides metabolism, Receptors, Purinergic P2 physiology
- Abstract
ADP-ribosylation is a post-translational modification regulating protein function in which amino acid-specific ADP-ribosyltransferases (ARTs) transfer ADP-ribose from NAD onto specific target proteins. Attachment of the bulky ADP-ribose usually inactivates the target by sterically blocking its interaction with other proteins. P2X7, an ATP-gated ion channel with important roles in inflammation and cell death, in contrast, is activated by ADP-ribosylation. Here, we report the structural basis for this gating and present the first molecular model for the activation of a target protein by ADP-ribosylation. We demonstrate that the ecto-enzyme ART2.2 ADP-ribosylates P2X7 at arginine 125 in a prominent, cysteine-rich region at the interface of 2 receptor subunits. ADP-ribose shares an adenine-ribonucleotide moiety with ATP. Our results indicate that ADP-ribosylation of R125 positions this common chemical framework to fit into the nucleotide-binding site of P2X7 and thereby gates the channel.
- Published
- 2008
- Full Text
- View/download PDF
47. Chemotaxis of mouse bone marrow neutrophils and dendritic cells is controlled by adp-ribose, the major product generated by the CD38 enzyme reaction.
- Author
-
Partida-Sanchez S, Gasser A, Fliegert R, Siebrands CC, Dammermann W, Shi G, Mousseau BJ, Sumoza-Toledo A, Bhagat H, Walseth TF, Guse AH, and Lund FE
- Subjects
- ADP-ribosyl Cyclase 1 deficiency, Adenosine Diphosphate Ribose chemical synthesis, Adenosine Diphosphate Ribose chemistry, Animals, Bone Marrow Cells immunology, Calcium antagonists & inhibitors, Calcium immunology, Cell Line, Chemotaxis immunology, Dendritic Cells drug effects, Dendritic Cells immunology, Leukocytes drug effects, Leukocytes immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, NAD analogs & derivatives, NAD pharmacology, Neutrophils immunology, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerases immunology, Sensitivity and Specificity, Structure-Activity Relationship, Time Factors, ADP-ribosyl Cyclase 1 metabolism, Adenosine Diphosphate Ribose analogs & derivatives, Adenosine Diphosphate Ribose pharmacology, Bone Marrow Cells drug effects, Chemotaxis drug effects, Neutrophils drug effects
- Abstract
The ectoenzyme CD38 catalyzes the production of cyclic ADP-ribose (cADPR) and ADP-ribose (ADPR) from its substrate, NAD(+). Both products of the CD38 enzyme reaction play important roles in signal transduction, as cADPR regulates calcium release from intracellular stores and ADPR controls cation entry through the plasma membrane channel TRPM2. We previously demonstrated that CD38 and the cADPR generated by CD38 regulate calcium signaling in leukocytes stimulated with some, but not all, chemokines and controls leukocyte migration to inflammatory sites. However, it is not known whether the other CD38 product, ADPR, also regulates leukocyte trafficking In this study we characterize 8-bromo (8Br)-ADPR, a novel compound that specifically inhibits ADPR-activated cation influx without affecting other key calcium release and entry pathways. Using 8Br-ADPR, we demonstrate that ADPR controls calcium influx and chemotaxis in mouse neutrophils and dendritic cells activated through chemokine receptors that rely on CD38 and cADPR for activity, including mouse FPR1, CXCR4, and CCR7. Furthermore, we show that the calcium and chemotactic responses of leukocytes are not dependent on poly-ADP-ribose polymerase 1 (PARP-1), another potential source of ADPR in some leukocytes. Finally, we demonstrate that NAD(+) analogues specifically block calcium influx and migration of chemokine-stimulated neutrophils without affecting PARP-1-dependent calcium responses. Collectively, these data identify ADPR as a new and important second messenger of mouse neutrophil and dendritic cell migration, suggest that CD38, rather than PARP-1, may be an important source of ADPR in these cells, and indicate that inhibitors of ADPR-gated calcium entry, such as 8Br-ADPR, have the potential to be used as anti-inflammatory agents.
- Published
- 2007
- Full Text
- View/download PDF
48. Modulation of Ca2+ entry and plasma membrane potential by human TRPM4b.
- Author
-
Fliegert R, Glassmeier G, Schmid F, Cornils K, Genisyuerek S, Harneit A, Schwarz JR, and Guse AH
- Subjects
- Animals, COS Cells, Calcium Channels genetics, Calcium Channels metabolism, Cell Line, Cell Membrane drug effects, Cell Membrane metabolism, Chlorocebus aethiops, Gene Expression, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Ionomycin pharmacology, Ionophores pharmacology, Membrane Potentials drug effects, Membrane Potentials genetics, Membrane Potentials physiology, Microscopy, Confocal, Patch-Clamp Techniques, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins physiology, Reverse Transcriptase Polymerase Chain Reaction, TRPM Cation Channels genetics, TRPM Cation Channels metabolism, Calcium metabolism, Calcium Channels physiology, Cell Membrane physiology, TRPM Cation Channels physiology
- Abstract
TRPM4b is a Ca(2+)-activated, voltage-dependent monovalent cation channel that has been shown to act as a negative regulator of Ca(2+) entry and to be involved in the generation of oscillations of Ca(2+) influx in Jurkat T-lymphocytes. Transient overexpression of TRPM4b as an enhanced green fluorescence fusion protein in human embryonic kidney (HEK) cells resulted in its localization in the plasma membrane, as demonstrated by confocal fluorescence microscopy. The functionality and plasma membrane localization of overexpressed TRPM4b was confirmed by induction of Ca(2+)-dependent inward and outward currents in whole cell patch clamp recordings. HEK-293 cells stably overexpressing TRPM4b showed higher ionomycin-activated Ca(2+) influx than wild-type cells. In addition, analysis of the membrane potential using the potentiometric dye bis-(1,3-dibutylbarbituric acid)-trimethine oxonol and by current clamp experiments in the perforated patch configuration revealed a faster initial depolarization after activation of Ca(2+) entry with ionomycin. Furthermore, TRPM4b expression facilitated repolarization and thereby enhanced sustained Ca(2+) influx. In conclusion, in cells with a small negative membrane potential, such as HEK-293 cells, TRPM4b acts as a positive regulator of Ca(2+) entry.
- Published
- 2007
- Full Text
- View/download PDF
49. Activation of T cell calcium influx by the second messenger ADP-ribose.
- Author
-
Gasser A, Glassmeier G, Fliegert R, Langhorst MF, Meinke S, Hein D, Krüger S, Weber K, Heiner I, Oppenheimer N, Schwarz JR, and Guse AH
- Subjects
- Cell Death, Concanavalin A pharmacology, Electrophysiology, Humans, Jurkat Cells, Lymphocyte Activation drug effects, TRPM Cation Channels analysis, TRPM Cation Channels genetics, TRPM Cation Channels metabolism, Adenosine Diphosphate Ribose metabolism, Adenosine Diphosphate Ribose physiology, Calcium metabolism, Second Messenger Systems physiology, T-Lymphocytes metabolism
- Abstract
Stimulation of Jurkat T cells by high concentrations of concanavalin A (ConA) induced an elevation of the endogenous adenosine diphosphoribose (ADPR) concentration and an inward current significantly different from the Ca2+ release-activated Ca2+ current (I(CRAC)). Electrophysiological characterization and activation of a similar current by infusion of ADPR indicated that the ConA-induced current is carried by TRPM2. Expression of TRPM2 in the plasma membrane of Jurkat T cells was demonstrated by reverse transcription-PCR, Western blot, and immunofluorescence. Inhibition of ADPR formation reduced ConA-mediated, but not store-operated, Ca2+ entry and prevented ConA-induced cell death of Jurkat cells. Moreover, gene silencing of TRPM2 abolished the ADPR- and ConA-mediated inward current. Thus, ADPR is a novel second messenger significantly involved in ConA-mediated cell death in T cells.
- Published
- 2006
- Full Text
- View/download PDF
50. Synthesis and biological evaluation of novel membrane-permeant cyclic ADP-ribose mimics: N1-[(5''-O-phosphorylethoxy)methyl]-5'-O-phosphorylinosine 5',5''-cyclicpyrophosphate (cIDPRE) and 8-substituted derivatives.
- Author
-
Gu X, Yang Z, Zhang L, Kunerth S, Fliegert R, Weber K, Guse AH, and Zhang L
- Subjects
- Calcium Signaling drug effects, Cell Membrane metabolism, Humans, Inosine Monophosphate chemistry, Inosine Monophosphate pharmacokinetics, Inosine Monophosphate pharmacology, Jurkat Cells, Molecular Mimicry, Permeability, Structure-Activity Relationship, Adenosine Diphosphate Ribose chemistry, Inosine Monophosphate analogs & derivatives, Inosine Monophosphate chemical synthesis
- Abstract
N1-[(5' '-O-Phosphorylethoxy)methyl]-5'-O-phosphorylinosine 5',5''-cyclicpyrophosphate (cIDPRE 2a) and the 8-substituted derivatives 8-bromo-, 8-azido-, 8-amino-, and 8-Cl-cIDPRE (2b-e) were synthesized from N1-[(5''-acetoxyethoxy)methyl]-2',3'-O-isopropylideneinosine (5) in good yields. The pharmacological activities of cIDPRE and the 8-substituted derivatives (2a-e) were analyzed in intact and permeabilized human Jurkat T-lymphocytes. The results indicate that cIDPRE permeates the plasma membrane, releases Ca2+ from an intracellular, cADPR-sensitive Ca2+ store, and subsequently initiates Ca2+ release-activated Ca2+ entry. The Ca(2+)-releasing activity of cIDPRE was confirmed directly in permeabilized cells. Using time-resolved confocal Ca2+ imaging at the single cell level, the development of global Ca2+ signals starting from local small Ca2+ signals evoked by cIDPRE was observed. 8-N3-cIDPRE 2c and 8-NH2-cIDPRE 2d were similarly effective in their agonistic activity as compared to cIDPRE 2a, showing almost indistinguishable concentration-response curves for 2a, 2c, and 2d and very similar kinetics of Ca2+ signaling. In contrast, the halogenated derivatives 8-Br- and 8-Cl-cIDPRE (2b and 2e) did not significantly elevate [Ca2+]i. Therefore, cIDPRE 2a, 8-N3-cIDPRE 2c, and 8-NH2-cIDPRE 2d are novel membrane permeant cADPR mimic and may provide important novel tools to study cADPR-mediated Ca2+ signaling in intact cells.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.