TY - JOUR AU - Csanády, László TI - Towards personalized medicine for cystic fibrosis patients with rare mutations JF - JOURNAL OF PHYSIOLOGY-LONDON J2 - J PHYSIOL-LONDON VL - 602 PY - 2024 IS - 2 SP - 257 EP - 258 PG - 2 SN - 0022-3751 DO - 10.1113/JP286135 UR - https://m2.mtmt.hu/api/publication/34507016 ID - 34507016 LA - English DB - MTMT ER - TY - JOUR AU - Szöllősi, András AU - Almássy, János TI - Functional characterization of the transient receptor potential melastatin 2 (TRPM2) cation channel from Nematostella vectensis reconstituted into lipid bilayer JF - SCIENTIFIC REPORTS J2 - SCI REP VL - 13 PY - 2023 IS - 1 PG - 10 SN - 2045-2322 DO - 10.1038/s41598-023-38640-6 UR - https://m2.mtmt.hu/api/publication/34064647 ID - 34064647 N1 - Department of Biochemistry, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary Department of Physiology, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary ELKH-SE Ion Channel Research Group, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary HCEMM-SE Molecular Channelopathies Research Group, Semmelweis University, Tuzolto u. 37-47, Budapest, 1094, Hungary Export Date: 4 September 2023 Correspondence Address: Szollosi, A.; Department of Biochemistry, Tuzolto u. 37-47, Hungary; email: szollo5@gmail.com Chemicals/CAS: adenosine diphosphate ribose, 20762-30-5; calcium, 7440-70-2, 14092-94-5; Adenosine Diphosphate Ribose; Calcium; Cations; Lipid Bilayers; TRPM Cation Channels Funding details: 739593 Funding details: Nemzeti Kutatási, Fejlesztési és Innovaciós Alap, NKFIA, KKP 144199 Funding text 1: This work was supported by Semmelweis Science and Innovation Fund STIA_18_M; EU Horizon 2020 Research and Innovation Program grant 739593 and National Research, Development and Innovation Fund grant KKP 144199. We thank László Csanády for funding and thorough reading and comments on the manuscript. We thank Balázs Tóth for help with poly- l -lysine measurements and stimulating discussions. AB - Transient receptor potential melastatin 2 (TRPM2) cation channel activity is required for insulin secretion, immune cell activation and body heat control. Channel activation upon oxidative stress is involved in the pathology of stroke and neurodegenerative disorders. Cytosolic Ca 2+ , ADP-ribose (ADPR) and phosphatidylinositol-4,5-bisphosphate (PIP 2 ) are the obligate activators of the channel. Several TRPM2 cryo-EM structures have been resolved to date, yet functionality of the purified protein has not been tested. Here we reconstituted overexpressed and purified TRPM2 from Nematostella vectensis (nvTRPM2) into lipid bilayers and found that the protein is fully functional. Consistent with the observations in native membranes, nvTRPM2 in lipid bilayers is co-activated by cytosolic Ca 2+ and either ADPR or ADPR-2′-phosphate (ADPRP). The physiological metabolite ADPRP has a higher apparent affinity than ADPR. In lipid bilayers nvTRPM2 displays a large linear unitary conductance, its open probability (P o ) shows little voltage dependence and is stable over several minutes. P o is high without addition of exogenous PIP 2 , but is largely blunted by treatment with poly- l -Lysine, a polycation that masks PIP 2 headgroups. These results indicate that PIP 2 or some other activating phosphoinositol lipid co-purifies with nvTRPM2, suggesting a high PIP 2 binding affinity of nvTRPM2 under physiological conditions. LA - English DB - MTMT ER - TY - JOUR AU - Csanády, László TI - Blue flash sheds light on the roles of individual phosphoserines in CFTR channel activation JF - JOURNAL OF GENERAL PHYSIOLOGY J2 - J GEN PHYSIOL VL - 155 PY - 2023 IS - 6 PG - 3 SN - 0022-1295 DO - 10.1085/jgp.202313336 UR - https://m2.mtmt.hu/api/publication/33755807 ID - 33755807 N1 - Department of Biochemistry, Semmelweis University, Budapest, Hungary HCEMM-SE Molecular Channelopathies Research Group, Budapest, Hungary ELKH-SE Ion Channel Research Group, Budapest, Hungary Export Date: 31 August 2023 CODEN: JEMEA Correspondence Address: Csanády, L.; Department of Biochemistry, Hungary; email: csanady.laszlo@med.semmelweis-univ.hu Chemicals/CAS: cyclic AMP, 60-92-4; cystic fibrosis transmembrane conductance regulator, 126880-72-6; phosphoserine, 407-41-0; serine, 56-45-1, 6898-95-9; Cystic Fibrosis Transmembrane Conductance Regulator; Phosphoserine; Serine Funding details: 739593 Funding details: Cystic Fibrosis Foundation, CFF, CSANAD21G0 Funding details: Nemzeti Kutatási Fejlesztési és Innovációs Hivatal, NKFIH, 144199 Funding text 1: This work was supported by EU Horizon 2020 Research and Innovation Program grant 739593, Cystic Fibrosis Foundation Research Grant CSANAD21G0, and Nemzeti Kutatási Fejlesztési és Innovációs Hivatal KKP_22 grant 144199 to L. Csanády. The author declares no competing financial interests. LA - English DB - MTMT ER - TY - JOUR AU - Simon, Márton AU - Csanády, László TI - Optimization of CFTR gating through the evolution of its extracellular loops JF - JOURNAL OF GENERAL PHYSIOLOGY J2 - J GEN PHYSIOL VL - 155 PY - 2023 IS - 4 PG - 16 SN - 0022-1295 DO - 10.1085/jgp.202213264 UR - https://m2.mtmt.hu/api/publication/33636409 ID - 33636409 LA - English DB - MTMT ER - TY - JOUR AU - Bartók, Ádám AU - Csanády, László TI - Dual amplification strategy turns TRPM2 channels into supersensitive central heat detectors JF - PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA J2 - P NATL ACAD SCI USA VL - 119 PY - 2022 IS - 48 PG - 11 SN - 0027-8424 DO - 10.1073/pnas.2212378119 UR - https://m2.mtmt.hu/api/publication/33262968 ID - 33262968 N1 - Hungarian Centre of Excellence for Molecular Medicine-Semmelweis Egyetem (HCEMM-SE), Molecular Channelopathies Research Group, Semmelweis University, Budapest, H-1094, Hungary Magyar Tudományos Akadémia-Semmelweis Egyetem (MTA-SE), Ion Channel Research Group, Semmelweis University, Budapest, H-1094, Hungary Department of Biochemistry, Semmelweis University, Budapest, H-1094, Hungary Cited By :3 Export Date: 4 September 2023 CODEN: PNASA Correspondence Address: Csanády, L.; Hungarian Centre of Excellence for Molecular Medicine-Semmelweis Egyetem (HCEMM-SE), Hungary; email: csanady.laszlo@med.semmelweis-univ.hu Chemicals/CAS: adenosine diphosphate ribose, 20762-30-5; calcium ion, 14127-61-8; hydrogen peroxide, 7722-84-1; calcium, 7440-70-2, 14092-94-5; Adenosine Diphosphate Ribose; Calcium; Hydrogen Peroxide; TRPM Cation Channels Funding details: 739593 Funding details: GINOP-2.3.2-15-2016-00051 Funding details: ÚNKP-20-5-SE-6, ÚNKP-21-5-SE-10 Funding details: Magyar Tudományos Akadémia, MTA, BO/00103/20, LP2017-14/2017 Funding details: Nemzeti Kutatási Fejlesztési és Innovációs Hivatal, NKFIH Funding text 1: We thank Beáta Töröcsik for subcloning T5L-TRPM2 into pcDNA3, Iordan Iordanov for providing purified nvNUDT9H, and Katalin Zboray (Agricultural Institute, Centre for Agricultural Research, Martonvásár, Hungary) for providing the stable T5L TRPM2 cell line (Economic Development and Innovation Operational Programme (GINOP-2.3.2-15-2016-00051) of the National Research, Development and Innovation Office). Support was provided by EU Horizon 2020 Research and Innovation Program grant 739593 and MTA Lendület grant LP2017-14/2017 to L.C. and a New National Excellence Program (ÚNKP) award of the Ministry of Human Capacities of Hungary to Semmelweis University. Á.B. was supported by the János Bolyai Research Scholarship of the Hungarian Academy of Sciences (BO/00103/20) and the New National Excellence Program (ÚNKP) Bolyai+ scholarship of the Ministry of Human Capacities of Hungary (ÚNKP-20-5-SE-6 and ÚNKP-21-5-SE-10). Funding text 2: ACKNOWLEDGMENTS. We thank Beáta Töröcsik for subcloning T5L-TRPM2 into pcDNA3, Iordan Iordanov for providing purified nvNUDT9H, and Katalin Zboray (Agricultural Institute, Centre for Agricultural Research, Martonvásár, Hungary) for providing the stableT5LTRPM2 cell line (Economic Development and Innovation Operational Programme (GINOP-2.3.2-15-2016-00051) of the National Research, Development and Innovation Office). Support was provided by EU Horizon 2020 Research and Innovation Program grant 739593 and MTA Lendület grant LP2017-14/2017 to L.C. and a New National Excellence Program (ÚNKP) award of the Ministry of Human Capacities of Hungary to Semmelweis University. Á.B. was supported by the János Bolyai Research Scholarship of the Hungarian Academy of Sciences (BO/00103/20) and the New National Excellence Program (ÚNKP) Bolyai+ scholarship of the Ministry of Human Capacities of Hungary (ÚNKP-20-5-SE-6 and ÚNKP-21-5-SE-10). AB - The Ca 2+ and ADP ribose (ADPR)-activated cation channel TRPM2 is the closest homolog of the cold sensor TRPM8 but serves as a deep-brain warmth sensor. To unravel the molecular mechanism of heat sensing by the TRPM2 protein, we study here temperature dependence of TRPM2 currents in cell-free membrane patches across ranges of agonist concentrations. We find that channel gating remains strictly agonist-dependent even at 40°C: heating alone or in combination with just Ca 2+ , just ADPR, Ca 2+ + cyclic ADPR, or H 2 O 2 pretreatment only marginally activates TRPM2. For fully liganded TRPM2, pore opening is intrinsically endothermic, due to ~10-fold larger activation enthalpy for opening (~200 kJ/mol) than for closure (~20 kJ/mol). However, the temperature threshold is too high (>40°C) for unliganded but too low (<15°C) for fully liganded channels. Thus, warmth sensitivity around 37°C is restricted to narrow ranges of agonist concentrations. For ADPR, that range matches, but for Ca 2+ , it exceeds bulk cytosolic values. The supraphysiological [Ca 2+ ] needed for TRPM2 warmth sensitivity is provided by Ca 2+ entering through the channel’s pore. That positive feedback provides further strong amplification to the TRPM2 temperature response (Q 10 ~ 1,000), enabling the TRPM2 protein to autonomously respond to tiny temperature fluctuations around 37°C. These functional data together with published structures suggest a molecular mechanism for opposite temperature dependences of two closely related channel proteins. LA - English DB - MTMT ER - TY - JOUR AU - Simon, Márton AU - Csanády, László TI - Molecular pathology of the R117H cystic fibrosis mutation is explained by loss of a hydrogen bond JF - ELIFE J2 - ELIFE VL - 10 PY - 2021 PG - 19 SN - 2050-084X DO - 10.7554/eLife.74693 UR - https://m2.mtmt.hu/api/publication/32573094 ID - 32573094 AB - The phosphorylation-activated anion channel cystic fibrosis transmembrane conductance regulator (CFTR) is gated by an ATP hydrolysis cycle at its two cytosolic nucleotide-binding domains, and is essential for epithelial salt-water transport. A large number of CFTR mutations cause cystic fibrosis. Since recent breakthrough in targeted pharmacotherapy, CFTR mutants with impaired gating are candidates for stimulation by potentiator drugs. Thus, understanding the molecular pathology of individual mutations has become important. The relatively common R117H mutation affects an extracellular loop, but nevertheless causes a strong gating defect. Here, we identify a hydrogen bond between the side chain of arginine 117 and the backbone carbonyl group of glutamate 1124 in the cryo-electronmicroscopic structure of phosphorylated, ATP-bound CFTR. We address the functional relevance of that interaction for CFTR gating using macroscopic and microscopic inside-out patch-clamp recordings. Employing thermodynamic double-mutant cycles, we systematically track gating-state-dependent changes in the strength of the R117-E1124 interaction. We find that the H-bond is formed only in the open state, but neither in the short-lived 'flickery' nor in the long-lived 'interburst' closed state. Loss of this H-bond explains the strong gating phenotype of the R117H mutant, including robustly shortened burst durations and strongly reduced intraburst open probability. The findings may help targeted potentiator design. LA - English DB - MTMT ER - TY - JOUR AU - Csanády, László TI - Degenerate but indispensable: How CFTR channel activity depends on the catalytically inactive ATP binding site JF - JOURNAL OF PHYSIOLOGY-LONDON J2 - J PHYSIOL-LONDON VL - 599 PY - 2021 IS - 20 SP - 4523 EP - 4524 PG - 2 SN - 0022-3751 DO - 10.1113/JP282278 UR - https://m2.mtmt.hu/api/publication/32358289 ID - 32358289 LA - English DB - MTMT ER - TY - JOUR AU - Szöllősi, András TI - Two decades of evolution of our understanding of the transient receptor potential melastatin 2 (Trpm2) cation channel JF - LIFE-BASEL J2 - LIFE-BASEL VL - 11 PY - 2021 IS - 5 PG - 23 SN - 2075-1729 DO - 10.3390/life11050397 UR - https://m2.mtmt.hu/api/publication/32040119 ID - 32040119 N1 - Department of Medical Biochemistry, Semmelweis University, Budapest, 1085, Hungary MTA-SE Lendület Ion Channel Research Group, Semmelweis University, Budapest, 1085, Hungary HCEMM-SE Molecular Channelopathies Research Group, Semmelweis University, Budapest, 1085, Hungary Cited By :2 Export Date: 9 March 2022 Correspondence Address: Szollosi, A.; Department of Medical Biochemistry, Hungary; email: szollosi.andras@med.semmelweis-univ.hu Funding details: 739593 Funding details: LP2017-14/2017 Funding text 1: Supported by MTA Lend?let grant LP2017-14/2017 and EU Horizon 2020 Research and Innovation Program grant 739593. Funding text 2: Funding: Supported by MTA Lendület grant LP2017-14/2017 and EU Horizon 2020 Research and Innovation Program grant 739593. AB - The transient receptor potential melastatin (TRPM) family belongs to the superfamily of TRP ion channels. It consists of eight family members that are involved in a plethora of cellular functions. TRPM2 is a homotetrameric Ca2+-permeable cation channel activated upon oxidative stress and is important, among others, for body heat control, immune cell activation and insulin secretion. Invertebrate TRPM2 proteins are channel enzymes; they hydrolyze the activating ligand, ADP-ribose, which is likely important for functional regulation. Since its cloning in 1998, the understanding of the biophysical properties of the channel has greatly advanced due to a vast number of structure– function studies. The physiological regulators of the channel have been identified and characterized in cell-free systems. In the wake of the recent structural biochemistry revolution, several TRPM2 cryo-EM structures have been published. These structures have helped to understand the general features of the channel, but at the same time have revealed unexplained mechanistic differences among channel orthologues. The present review aims at depicting the major research lines in TRPM2 structure-function. It discusses biophysical properties of the pore and the mode of action of direct channel effectors, and interprets these functional properties on the basis of recent three-dimensional structural models. © 2021 by the author. Licensee MDPI, Basel, Switzerland. LA - English DB - MTMT ER - TY - JOUR AU - Mihályi, Csaba AU - Iordanov, Iordan AU - Törőcsik, Beáta AU - Csanády, László TI - Simple binding of protein kinase A, prior to phosphorylation, allows CFTR anion channels to be opened by nucleotides JF - PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA J2 - P NATL ACAD SCI USA VL - 117 PY - 2020 IS - 35 SP - 21740 EP - 21746 PG - 7 SN - 0027-8424 DO - 10.1073/pnas.2007910117 UR - https://m2.mtmt.hu/api/publication/31397038 ID - 31397038 LA - English DB - MTMT ER - TY - JOUR AU - Tóth, Balázs AU - Iordanov, Iordan AU - Csanády, László TI - Selective profiling of N- And C-terminal nucleotide-binding sites in a TRPM2 channel JF - JOURNAL OF GENERAL PHYSIOLOGY J2 - J GEN PHYSIOL VL - 152 PY - 2020 IS - 5 PG - 13 SN - 0022-1295 DO - 10.1085/jgp.201912533 UR - https://m2.mtmt.hu/api/publication/31365406 ID - 31365406 N1 - Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary MTA-SE Lendület Ion Channel Research Group, Semmelweis University, Budapest, Hungary Cited By :7 Export Date: 15 September 2021 CODEN: JGPLA Correspondence Address: Tóth, B.; Department of Medical Biochemistry, Hungary; email: toth.balazs@med.semmelweis-univ.hu Chemicals/CAS: adenosine diphosphate ribose, 20762-30-5; amino acid, 65072-01-7; calcium, 7440-70-2, 14092-94-5; Adenosine Diphosphate Ribose; Amino Acids; Calcium; Ligands; Nucleotides; TRPM Cation Channels; TRPM2 protein, human Funding details: Semmelweis Egyetem, 18-4-SE-132, 19-4-SE-49 Funding details: Magyar Tudományos Akadémia, MTA, LP2017-14/2017 Funding text 1: Supported by the Hungarian Academy of Sciences (Lendület grant LP2017-14/2017 to L. Csanády) and a Ministry of Human Capacities of Hungary New National Excellence Program (Új Nemzeti Kiválóság Program) award to Semmelweis University. B. Tóth is a János Bolyai Research Fellow, supported by postdoctoral Új Nemzeti Kiválóság Program grants 18-4-SE-132 and 19-4-SE-49. The authors declare no competing financial interests. AB - Transient receptor potential melastatin 2 (TRPM2) is a homotetrameric Ca2+-permeable cation channel important for the immune response, body temperature regulation, and insulin secretion, and is activated by cytosolic Ca2+ and ADP ribose (ADPR). ADPR binds to two distinct locations, formed by large N- and C-terminal cytosolic domains, respectively, of the channel protein. In invertebrate TRPM2 channels, the C-terminal site is not required for channel activity but acts as an active ADPR phosphohydrolase that cleaves the activating ligand. In vertebrate TRPM2 channels, the C-terminal site is catalytically inactive but cooperates with the N-terminal site in channel activation. The precise functional contributions to channel gating and the nucleotide selectivities of the two sites in various species have not yet been deciphered. For TRPM2 of the sea anemone Nematostella vectensis (nvTRPM2), catalytic activity is solely attributable to the C-terminal site. Here, we show that nvTRPM2 channel gating properties remain unaltered upon deletion of the C-terminal domain, indicating that the N-terminal site is single-handedly responsible for channel gating. Exploiting such functional independence of the N- and C-terminal sites, we selectively measure their affinity profiles for a series of ADPR analogues, as reflected by apparent affinities for channel activation and catalysis, respectively. Using site-directed mutagenesis, we confirm that the same N-terminal site observed in vertebrate TRPM2 channels was already present in ancient cnidarians. Finally, by characterizing the functional effects of six amino acid side chain truncations in the N-terminal site, we provide first insights into the mechanistic contributions of those side chains to TRPM2 channel gating. © 2020 Tóth et al. This article is distributed under the terms of an Attribution-Noncommercial-Share Alike-No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms/). After six months it is available under a Creative Commons License (Attribution-Noncommercial-Share Alike 4.0 International license, as described at https://creativecommons.org/licenses/by-nc-sa/4.0/). LA - English DB - MTMT ER -