1
|
Adaikalapandi S, Thangadurai TD, Sivakumar S, Nataraj D, Schechter A, Kalarikkal N, Thomas S. Aggregation induced emission "Turn on" ultra-low detection of anti-inflammatory drug flufenamic acid in human urine samples by carbon dots derived from bamboo stem waste. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 326:125278. [PMID: 39423556 DOI: 10.1016/j.saa.2024.125278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Carbon dot-based fluorescence sensors have attracted research interest for the selective determination of anti-inflammatory drugs in biological fluids and environments. The overdose and accumulation of anti-inflammatory drugs in tissues can cause chronic side effects including abdominal pain, and renal damage. Herein, we report a new fluorescent probe, bamboo stem waste-derived carbon dots (BS-CDs) for highly sensitive detection of Flufenamic acid (FA), a hazardous anti-inflammatory drug. The UV-vis absorption spectra of BS-CDs show a redshifted absorption peak at 283 nm upon the addition of FA suggesting strong binding interaction between BS-CDs and FA molecule. The BS-CDs showed a fluorescence enhancement (∼2-fold) detection for FA (400 μM) in the linear concentration range (0.40 → 0.65 μM) with a limit of detection (LoD; 17 nM) and binding constant (Ka = 1.33 × 10-3 M-1). The time-resolved fluorescence decay analysis showed that the average lifetime of BS-CDs has slightly changed (4.42 → 4.67 ns) by the interaction with FA through the aggregation-induced emission (AIE) process. The interference, pH, and effect of time results suggest that BS-CDs are highly selective probes for FA detection and do not show any interference involvement during FA detection. The confirmation of the structure and morphology changes of BS-CDs after interaction with FA was carried out by XRD, FESEM, HRTEM, FTIR, and Raman spectroscopy. The practicability of the BS-CDs probe was proved by the detection of FA in human urine samples with recovery of 103-109 %. This suggests that the proposed BS-CDs-based 'turn-on' sensor could be used to determine the FA in biological fluids.
Collapse
Affiliation(s)
- Subitha Adaikalapandi
- Department of Chemistry, and Centre for Research and Development (CFRD), KPR Institute of Engineering and Technology, Coimbatore, 641407, Tamil Nadu, India
| | - T Daniel Thangadurai
- Department of Chemistry, and Centre for Research and Development (CFRD), KPR Institute of Engineering and Technology, Coimbatore, 641407, Tamil Nadu, India.
| | - S Sivakumar
- Department of Chemistry, and Centre for Research and Development (CFRD), KPR Institute of Engineering and Technology, Coimbatore, 641407, Tamil Nadu, India
| | - D Nataraj
- Department of Physics, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Alex Schechter
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel
| | - Nandakumar Kalarikkal
- School of Pure and Applied Physics, Mahatma Gandhi University, Kottayam 686560, Kerala, India
| | - Sabu Thomas
- School of Chemical Sciences, Mahatma Gandhi University, Kottayam 686560, Kerala, India
| |
Collapse
|
2
|
Li S, Xuan B, Wong SN, Lee HW, Low KH, Chow SF. Towards the discovery of unrevealed flufenamic acid cocrystals via structural resemblance for enhanced topical drug delivery. Int J Pharm 2025; 668:124959. [PMID: 39550015 DOI: 10.1016/j.ijpharm.2024.124959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/22/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Cocrystallization has emerged as a promising formulation strategy for modulating transdermal drug absorption by enhancing solubility and permeability. However, challenges related to cocrystal dissociation in the semi-solid state need to be addressed to mitigate regulatory concerns before the widespread implementation of topical cocrystal products in clinical practice. This study aimed to develop oil-based topical formulations incorporating cocrystals with distinct thermodynamic stabilities, followed by investigating the roles of different structurally similar coformers and oily vehicles on their physicochemical properties. Three pharmaceutical cocrystals of poorly water-soluble flufenamic acid (FFA) were synthesized with isomeric pyridine carboxamides in a 1:1 stoichiometry via rapid solvent removal. These included the reported flufenamic acid-nicotinamide cocrystal (FFA-NIC), the long-elusive flufenamic acid-isonicotinamide cocrystal (FFA-IST) and flufenamic acid-picolinamide cocrystal (FFA-PIC). The resulting cocrystals, which exhibited different hydrogen bonding patterns, were characterized using powder X-ray diffraction, differential scanning calorimetry, Fourier transform infrared spectroscopy, and structural analysis through single crystal X-ray diffraction. The cocrystals were further formulated in a series of oleaginous and absorption bases, including liquid paraffin, Vaseline, lanolin, and theobroma oil, for topical delivery. The cocrystal dissociation, content uniformity, and in vitro membrane diffusion were assessed. Notably, although all FFA cocrystals exhibited thermodynamic instability in aqueous solution, a significantly reduced propensity for cocrystal dissociation was observed in the ointment bases. Integrated computational analyses of packing efficiency and interaction energy revealed that the thermodynamic stability of cocrystals followed a descending order of FFA-NIC > FFA-PIC > FFA-IST. Compared with raw FFA, FFA-IST and FFA-PIC, which had larger positive ΔVnon-H and ΔEcocryst, achieved superior cumulative diffusion of FFA from Vaseline, with a 4.3-fold (p = 0.0003) and 3.3-fold (p = 0.0029) increase at 6 h in a Franz diffusion cell model, respectively. The diffusion of all FFA cocrystals mainly followed the Higuchi kinetic model and was positively correlated with the intrinsic dissolution rate.
Collapse
Affiliation(s)
- Si Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Bianfei Xuan
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Si Nga Wong
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Hok Wai Lee
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kam-Hung Low
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Shing Fung Chow
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong.
| |
Collapse
|
3
|
Binkle-Ladisch L, Pironet A, Zaliani A, Alcouffe C, Mensching D, Haferkamp U, Willing A, Woo MS, Erdmann A, Jessen T, Hess SD, Gribbon P, Pless O, Vennekens R, Friese MA. Identification and development of TRPM4 antagonists to counteract neuronal excitotoxicity. iScience 2024; 27:111425. [PMID: 39687019 PMCID: PMC11648915 DOI: 10.1016/j.isci.2024.111425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration in central nervous system disorders is linked to dysregulated neuronal calcium. Direct inhibition of glutamate-induced neuronal calcium influx, particularly via N-methyl-D-aspartate receptors (NMDAR), has led to adverse effects and clinical trial failures. A more feasible approach is to modulate NMDAR activity or calcium signaling indirectly. In this respect, the calcium-activated non-selective cation channel transient receptor potential melastatin 4 (TRPM4) has been identified as a promising target. However, high affinity and specific antagonists are lacking. Here, we conducted high-throughput screening of a compound library to identify high affinity TRPM4 antagonists. This yielded five lead compound series with nanomolar half-maximal inhibitory concentration values. Through medicinal chemistry optimization of two series, we established detailed structure-activity relationships and inhibition of excitotoxicity in neurons. Moreover, we identified their potential binding site supported by electrophysiological measurements. These potent TRPM4 antagonists are promising drugs for treating neurodegenerative disorders and TRPM4-related pathologies, potentially overcoming previous therapeutic challenges.
Collapse
Affiliation(s)
- Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Chantal Alcouffe
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | - Daniel Mensching
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcel S. Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Alexandre Erdmann
- Department of Chemistry, Evotec SE, 195 Route D'Espagne, 31036 Toulouse, France
| | | | - Stephen D. Hess
- Evotec Asia Pte Ltd, 79 Science Park Drive, #04-05 Cintech IV, Singapore 118264, Singapore
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Campus Gasthuisberg O/N1, Herestraat 49-Bus 802, 3000 Leuven, Belgium
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
4
|
Sinyak DS, Amakhin DV, Soboleva EB, Gryaznova MO, Zaitsev AV. Flufenamic acid abolishes epileptiform activity in the entorhinal cortex slices by reducing the temporal summation of glutamatergic responses. Biochem Biophys Res Commun 2024; 733:150666. [PMID: 39244848 DOI: 10.1016/j.bbrc.2024.150666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Flufenamic acid (FFA) is an anti-inflammatory drug that affects multiple targets and is a widely used research tool in ion channel studies. This pharmacological compound has a low level of selectivity for the transient receptor potential (TRP) channel superfamily, blocking calcium-activated nonselective cation current (ICAN) as well as afterdepolarizations (ADP) induced by it. A number of studies have demonstrated that FFA exerts an anti-epileptic effect in vitro, although the precise mechanism of this effect is not yet identified. The present study used whole-cell patch-clamp recordings and demonstrated that FFA (25 μM) can abolish the generation of seizure-like events (SLE) in entorhinal cortex slices perfused with a 4-aminopyridine-containing solution, depending on the time of application. FFA decreased the temporal summation of synaptic potentials at the onset of SLEs. However, as the epileptiform activity evolved and the SLE onset phase became more abrupt, the blocking effect of FFA diminished. FFA effectively abolished TRP channel-mediated slow ADPs, exerted a weak blockade and slowed the kinetics of GABAa receptor-mediated currents, and did not affect NMDA receptor-mediated evoked currents induced by extracellular stimulation. Although FFA did not directly inhibit NMDA receptor-mediated evoked currents, it decreased the summation of NMDA receptor-mediated potentials in a manner comparable to its effect on the initiation phase of SLE. This suggests that ICAN blockade may be responsible for this effect. Furthermore, our results showed that the selective blocker of melastatin TRP channels (TRPM4) 9-phenanthrol effectively abolished epileptiform activity in a manner analogous to FFA. In contrast, ML-204, the blocker of canonical TRP channels (TRPC), had no discernible effect on this phenomenon. In conclusion, the study demonstrate that FFA abolishes epileptiform activity in the entorhinal cortex by blocking TRPM4 channels and, consequently, decreasing the effectiveness of temporal summation of glutamatergic potentials.
Collapse
Affiliation(s)
- Denis S Sinyak
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, pr. Torez 44, 194223, Saint Petersburg, Russia
| | - Dmitry V Amakhin
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, pr. Torez 44, 194223, Saint Petersburg, Russia
| | - Elena B Soboleva
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, pr. Torez 44, 194223, Saint Petersburg, Russia
| | - Marusya O Gryaznova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, pr. Torez 44, 194223, Saint Petersburg, Russia
| | - Aleksey V Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, pr. Torez 44, 194223, Saint Petersburg, Russia.
| |
Collapse
|
5
|
Ying K, Hua N, Luo Y, Liu X, Liu M, Yang W. [Construction of HEK293T cell line stably expressing TRPM2 channel based on PiggyBac transposition system and its application in drug screening for cerebral ischemia and other diseases]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:604-614. [PMID: 39343750 PMCID: PMC11528149 DOI: 10.3724/zdxbyxb-2024-0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES To establish a cell line stably expressing the transient receptor potential melastatin 2 (TRPM2) channel for screening TRPM2 inhibitors based on PiggyBac transposition system. METHODS A plasmid PiggyBac-human TRPM2 (pPB-hTRPM2) eukaryotic expression vector was constructed using PiggyBac transposition system. The plasmid and a helper plasmid were co-transfected into HEK293T cells to express TRPM2, which was identified by fluorescence and patch-clamp assays. The high throughput screening performance was assessed with the Z ´ factor. Calcium imaging and patch clamp techniques were employed to assess the initial activity of eleven compound molecules, confirming the inhibitory effects of the primary molecules on TRPM2. The protective effect of the screened compounds on damaged cells was validated using the oxygen-glucose deprivation/reperfusion (OGD/R) injury model and CCK-8 kit. The level of cellular reactive oxygen species (ROS) was detected by flow cytometry. The neuroprotective effects of the compounds were evaluated using a transient middle cerebral artery occlusion (tMCAO) mouse model. RESULTS The HEK293T cells transfected with pPB-hTRPM2-EGFP showed high TRPM2 expression. Puromycin-resistant cells, selected through screening, exhibited robust fluorescence. Whole-cell patch results revealed that induced cells displayed classical TRPM2 current characteristics comparable to the control group, showing no significant differences (P>0.05). With a Z ´ factor of 0.5416 in calcium imaging, the model demonstrated suitability for high-throughput screening of TRPM2 inhibitors. Calcium imaging and electrophysiological experiments indicated that compound 6 significantly inhibited the TRPM2 channel. Further experiments showed that 1.0 μmol/L of compound 6 enhanced cell viability (P<0.05) and reduced the level of ROS (P<0.05) of SH-SY5Y under OGD/R injury. 0.3 and 1.0 mg/kg of compound 6 reduced the cerebral infarction volume in tMCAO mice (both P<0.05). CONCLUSIONS A stable TRPM2 gene expressing cell line has been successfully established using PiggyBac gene editing in this study. TRPM2 channel inhibitors were screened through calcium imaging and patch clamp techniques, and an inhibitor compound 6 was identified. This compound can alleviate cell damage after OGD/R by reducing cellular ROS levels and has a protective effect against cerebral ischemia-reperfusion injury in mice.
Collapse
Affiliation(s)
- Kaiyue Ying
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Ning Hua
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanping Luo
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xingyu Liu
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Min Liu
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Yang
- Department of Biophysics, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
6
|
Guo Y, Cheng D, Yu ZY, Schiatti T, Chan AY, Hill AP, Peyronnet R, Feneley MP, Cox CD, Martinac B. Functional coupling between Piezo1 and TRPM4 influences the electrical activity of HL-1 atrial myocytes. J Physiol 2024; 602:4363-4386. [PMID: 38098265 DOI: 10.1113/jp284474] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 11/30/2023] [Indexed: 09/25/2024] Open
Abstract
The transient receptor potential melastatin 4 (TRPM4) channel contributes extensively to cardiac electrical activity, especially cardiomyocyte action potential formation. Mechanical stretch can induce changes in heart rate and rhythm, and the mechanosensitive channel Piezo1 is expressed in many cell types within the myocardium. Our previous study showed that TRPM4 and Piezo1 are closely co-localized in the t-tubules of ventricular cardiomyocytes and contribute to the Ca2+-dependent signalling cascade that underlies hypertrophy in response to mechanical pressure overload. However, there was no direct evidence showing that Piezo1 activation was related to TRPM4 activation in situ. In the present study, we employed the HL-1 mouse atrial myocyte-like cell line as an in vitro model to investigate whether Piezo1-TRPM4 coupling can affect action potential properties. We used the small molecule Piezo1 agonist, Yoda1, as a surrogate for mechanical stretch to activate Piezo1 and detected the action potential changes in HL-1 cells using FluoVolt, a fluorescent voltage sensitive dye. Our results demonstrate that Yoda1-induced activation of Piezo1 changes the action potential frequency in HL-1 cells. This change in action potential frequency is reduced by Piezo1 knockdown using small intefering RNA. Importantly knockdown or pharmacological inhibition of TRPM4 significantly affected the degree to which Yoda1-evoked Piezo1 activation influenced action potential frequency. Thus, the present study provides in vitro evidence of a functional coupling between Piezo1 and TRPM4 in a cardiomyocyte-like cell line. The coupling of a mechanosensitive Ca2+ permeable channel and a Ca2+-activated TRP channel probably represents a ubiquitous model for the role of TRP channels in mechanosensory transduction. KEY POINTS: The transient receptor potential melastatin 4 (TRPM4) and Piezo1 channels have been confirmed to contribute to the Ca2+-dependent signalling cascade that underlies cardiac hypertrophy in response to mechanical pressure overload. However, there was no direct evidence showing that Piezo1 activation was related to TRPM4 activation in situ. We employed the HL-1 mouse atrial myocyte-like cell line as an in vitro model to investigate the effect of Piezo1-TRPM4 coupling on cardiac electrical properties. The results show that both pharmacological and genetic inhibition of TRPM4 significantly affected the degree to which Piezo1 activation influenced action potential frequency in HL-1 cells. Our findings provide in vitro evidence of a functional coupling between Piezo1 and TRPM4 in a cardiomyocyte-like cell line. The coupling of a mechanosensitive Ca2+ permeable channel and a Ca2+-activated TRP channel probably represents a ubiquitous model for the role of TRP channels in mechanosensory transduction in various (patho)physiological processes.
Collapse
Affiliation(s)
- Yang Guo
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Delfine Cheng
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Ze-Yan Yu
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Teresa Schiatti
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrea Y Chan
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael P Feneley
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Cardiology, St Vincent's Hospital, Sydney, NSW, Australia
| | - Charles D Cox
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
7
|
Kasuya K, Takahashi K, Hashimoto M, Ohta T. Nociceptive transient receptor potential ankyrin 1 (TRPA1) in sensory neurons are targets of the antifungal drug econazole. BMC Pharmacol Toxicol 2024; 25:53. [PMID: 39169383 PMCID: PMC11337588 DOI: 10.1186/s40360-024-00779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Econazole is a widely used imidazole derivative antifungal for treating skin infections. The molecular targets for its frequent adverse effects of skin irritation symptoms, such as pruritus, burning sensation, and pain, have not been clarified. Transient receptor potential (TRP) channels, non-selective cation channels, are mainly expressed in peripheral sensory neurons and serve as sensors for various irritants. METHODS We investigated the effect of econazole on TRP channel activation by measuring intracellular calcium concentration ([Ca2+]i) through fluorescent ratio imaging in mouse dorsal root ganglion (DRG) neurons isolated from wild-type, TRPA1(-/-) and TRPV1(-/-) mice, as well as in heterologously TRP channel-expressed cells. A cheek injection model was employed to assess econazole-induced itch and pain in vivo. RESULTS Econazole evoked an increase in [Ca2+]i, which was abolished by the removal of extracellular Ca2+ in mouse DRG neurons. The [Ca2+]i responses to econazole were suppressed by a TRPA1 blocker but not by a TRPV1 blocker. Attenuation of the econazole-induced [Ca2+]i responses was observed in the TRPA1(-/-) mouse DRG neurons but was not significant in the TRPV1(-/-) neurons. Econazole increased the [Ca2+]i in HEK293 cells expressing TRPA1 (TRPA1-HEK) but not in those expressing TRPV1, although at higher concentrations, it induced Ca2+ mobilization from intracellular stores in untransfected naïve HEK293 cells. Miconazole, which is a structural analog of econazole, also increased the [Ca2+]i in mouse DRG neurons and TRPA1-HEK, and its nonspecific action was larger than econazole. Fluconazole, a triazole drug failed to activate TRPA1 and TRPV1 in mouse DRG neurons and TRPA1-HEK. Econazole induced itch and pain in wild-type mice, with reduced responses in TRPA1(-/-) mice. CONCLUSIONS These findings suggested that the imidazole derivatives econazole and miconazole may induce skin irritation by activating nociceptive TRPA1 in the sensory neurons. Suppression of TRPA1 activation may mitigate the adverse effects of econazole.
Collapse
Affiliation(s)
- Kaoru Kasuya
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Kenji Takahashi
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
- Department of Veterinary Pharmacology, Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan
| | - Miho Hashimoto
- Department of Veterinary Pharmacology, Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan
| | - Toshio Ohta
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan.
- Department of Veterinary Pharmacology, Joint Graduate School of Veterinary Sciences, Tottori University, Tottori, 680-8553, Japan.
| |
Collapse
|
8
|
Salmanzadeh H, Halliwell RF. Antiseizure properties of fenamate NSAIDs determined in mature human stem-cell derived neuroglial circuits. Front Pharmacol 2024; 15:1385523. [PMID: 38828453 PMCID: PMC11141243 DOI: 10.3389/fphar.2024.1385523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024] Open
Abstract
Repeated and uncontrolled seizures in epilepsy result in brain cell loss and neural inflammation. Current anticonvulsants primarily target ion channels and receptors implicated in seizure activity. Identification of neurotherapeutics that can inhibit epileptiform activity and reduce inflammation in the brain may offer significant benefits in the long-term management of epilepsy. Fenamates are unique because they are both non-steroidal anti-inflammatory drugs (NSAIDs) and highly subunit selective modulators of GABAA receptors. In the current study we have investigated the hypothesis that fenamates have antiseizure properties using mature human stem cell-derived neuro-glia cell cultures, maintained in long-term culture, and previously shown to be sensitive to first, second and third generation antiepileptics. Mefenamic acid, flufenamic acid, meclofenamic acid, niflumic acid, and tolfenamic acid (each tested at 10-100 μM) attenuated 4-aminopyridine (4-AP, 100 μM) evoked epileptiform activity in a dose-dependent fashion. These actions were as effective diazepam (3-30 μM) and up to 200 times more potent than phenobarbital (300-1,000 μM). The low (micromolar) concentrations of fenamates that inhibited 4-AP evoked epileptiform activity correspond to those reported to potentiate GABAA receptor function. In contrast, the fenamates had no effect on neural spike amplitudes, indicating that their antiseizure actions did not result from inhibition of sodium-channels. The antiseizure actions of fenamates were also not replicated by either of the two non-fenamate NSAIDs, ibuprofen (10-100 μM) or indomethacin (10-100 μM), indicating that inhibition of cyclooxygenases is not the mechanism through which fenamates have anticonvulsant properties. This study therefore shows for the first time, using functionally mature human stem cell-derived neuroglial circuits, that fenamate NSAIDs have powerful antiseizure actions independent of, and in addition to their well-established anti-inflammatory properties, suggesting these drugs may provide a novel insight and new approach to the treatment of epilepsy in the future.
Collapse
Affiliation(s)
| | - Robert F. Halliwell
- Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States
| |
Collapse
|
9
|
Morse J, Wang D, Mei S, Whitham D, Hladun C, Darie CC, Sintim HO, Wang M, Leung K. Chloride Homeostasis Regulates cGAS-STING Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588475. [PMID: 38645072 PMCID: PMC11030317 DOI: 10.1101/2024.04.08.588475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The cGAS-STING signaling pathway has emerged as a key mediator of inflammation. However, the roles of chloride homeostasis on this pathway are unclear. Here, we uncovered a correlation between chloride homeostasis and cGAS-STING signaling. We found that dysregulation of chloride homeostasis attenuates cGAS-STING signaling in a lysosome-independent manner. Treating immune cells with chloride channel inhibitors attenuated 2'3'-cGAMP production by cGAS and also suppressed STING polymerization, leading to reduced cytokine production. We also demonstrate that non-selective chloride channel blockers can suppress the NPC1 deficiency-induced, hyper-activated STING signaling in skin fibroblasts derived from Niemann Pick disease type C (NPC) patients. Our findings reveal that chloride homeostasis majorly affects cGAS-STING pathway and suggest a provocative strategy to dampen STING-mediated inflammation via targeting chloride channels.
Collapse
Affiliation(s)
- Jared Morse
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danna Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Serena Mei
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danielle Whitham
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Colby Hladun
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Costel C. Darie
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Modi Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - KaHo Leung
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| |
Collapse
|
10
|
Kappel S, Melek K, Ross-Kaschitza D, Hauert B, Gerber CE, Lochner M, Peinelt C. CBA (4-chloro-2-(2-chlorophenoxy)acetamido) benzoic acid) inhibits TMEM206 mediated currents and TMEM206 does not contribute to acid-induced cell death in colorectal cancer cells. Front Pharmacol 2024; 15:1369513. [PMID: 38515848 PMCID: PMC10955468 DOI: 10.3389/fphar.2024.1369513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction: Upon activation at low pH, TMEM206 conducts Cl- ions across plasma and vesicular membranes. In a (patho)physiological context, TMEM206 was reported to contribute to acid-induced cell death in neurons, kidney and cervical epithelial cells. We investigated the role of TMEM206 in acid-induced cell death in colorectal cancer cells. In addition, we studied CBA as a new small molecule inhibitor for TMEM206. Methods: The role of TMEM206 in acid-induced cell death was studied with CRISPR/Cas9-mediated knockout and FACS analysis. The pharmacology of TMEM206 was determined with the patch clamp technique. Results: In colorectal cancer cells, TMEM206 is not a critical mediator of acid-induced cell death. CBA is a small molecule inhibitor of TMEM206 (IC50 = 9.55 µM) at low pH, at pH 6.0 inhibition is limited. Conclusion: CBA demonstrates effective and specific inhibition of TMEM206; however, its inhibitory efficacy is limited at pH 6.0. Despite this limitation, CBA is a potent inhibitor for functional studies at pH 4.5 and may be a promising scaffold for the development of future TMEM206 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Kapuścińska D, Narajczyk M, Liakh I, Wielgomas B, Aksmann A. Nabumetone and flufenamic acid pose a serious risk to aquatic plants: A study with Chlamydomonas reinhardtii as a model organism. CHEMOSPHERE 2024; 349:140853. [PMID: 38052310 DOI: 10.1016/j.chemosphere.2023.140853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/25/2023] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
The aquatic environment is constantly under threat due to the release of numerous pollutants. Among them, pharmaceuticals constitute a huge and diverse group. Non-steroidal anti-inflammatory drugs (NSAIDs) are increasingly found in water bodies, but knowledge about their potential toxicity is still low. In particular, there is a lack of information about their influences on aquatic plants and algae. We estimated the susceptibility of the microalgae Chlamydomonas reinhardtii to nabumetone (NBT) and flufenamic acid (FFA), focusing on photosynthesis. Due to the differences in the structures of these compounds, it was assumed that these drugs would have different toxicities to the tested green algae. The hypothesis was confirmed by determining the effective concentration values, the intensity of photosynthesis, the intensity of dark respiration, the contents of photosynthetic pigments, the fluorescence of chlorophyll a in vivo (OJIP test), and cell ultrastructure analysis. Assessment of the toxicity of the NSAIDs was extended by the calculation of an integrated biomarker response index (IBR), which is a valuable tool in ecotoxicological studies. The obtained results indicate an over six times higher toxicity of NBT compared to FFA. After analysis of the chlorophyll a fluorescence in vivo, it was found that NBT inhibited electron transport beyond the PS II. FFA, unlike NBT, lowered the intensity of photosynthesis, probably transforming some reaction centers into "silent centers", which dissipate energy as heat. The IBR estimated based on photosynthetic parameters suggests that the toxic effect of FFA results mainly from photosynthesis disruption, whereas NBT significantly affects other cellular processes. No significant alteration in the ultrastructure of treated cells could be seen, except for changes in starch grain number and autophagic vacuoles that appeared in FFA-treated cells. To the best of our knowledge, this is the first work reporting the toxic effects of NBT and FFA on unicellular green algae.
Collapse
Affiliation(s)
- Dominika Kapuścińska
- Department of Plant Experimental Biology and Biotechnology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland.
| | - Magdalena Narajczyk
- Laboratory of Electron Microscopy, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308, Gdansk, Poland.
| | - Ivan Liakh
- Department of Toxicology, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland.
| | - Bartosz Wielgomas
- Department of Toxicology, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland.
| | - Anna Aksmann
- Department of Plant Experimental Biology and Biotechnology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland.
| |
Collapse
|
12
|
Monteil A, Guérineau NC, Gil-Nagel A, Parra-Diaz P, Lory P, Senatore A. New insights into the physiology and pathophysiology of the atypical sodium leak channel NALCN. Physiol Rev 2024; 104:399-472. [PMID: 37615954 DOI: 10.1152/physrev.00014.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cell excitability and its modulation by hormones and neurotransmitters involve the concerted action of a large repertoire of membrane proteins, especially ion channels. Unique complements of coexpressed ion channels are exquisitely balanced against each other in different excitable cell types, establishing distinct electrical properties that are tailored for diverse physiological contributions, and dysfunction of any component may induce a disease state. A crucial parameter controlling cell excitability is the resting membrane potential (RMP) set by extra- and intracellular concentrations of ions, mainly Na+, K+, and Cl-, and their passive permeation across the cell membrane through leak ion channels. Indeed, dysregulation of RMP causes significant effects on cellular excitability. This review describes the molecular and physiological properties of the Na+ leak channel NALCN, which associates with its accessory subunits UNC-79, UNC-80, and NLF-1/FAM155 to conduct depolarizing background Na+ currents in various excitable cell types, especially neurons. Studies of animal models clearly demonstrate that NALCN contributes to fundamental physiological processes in the nervous system including the control of respiratory rhythm, circadian rhythm, sleep, and locomotor behavior. Furthermore, dysfunction of NALCN and its subunits is associated with severe pathological states in humans. The critical involvement of NALCN in physiology is now well established, but its study has been hampered by the lack of specific drugs that can block or agonize NALCN currents in vitro and in vivo. Molecular tools and animal models are now available to accelerate our understanding of how NALCN contributes to key physiological functions and the development of novel therapies for NALCN channelopathies.
Collapse
Affiliation(s)
- Arnaud Monteil
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Antonio Gil-Nagel
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Paloma Parra-Diaz
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
13
|
Chen Z, Cheng Z, Ding C, Cao T, Chen L, Wang H, Li J, Huang X. ROS-Activated TRPM2 Channel: Calcium Homeostasis in Cardiovascular/renal System and Speculation in Cardiorenal Syndrome. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07531-3. [PMID: 38108918 DOI: 10.1007/s10557-023-07531-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
The transient receptor potential melastatin 2 (TRPM2) channel is a nonselective calcium channel that is sensitive to oxidative stress (OS), and is widely expressed in multiple organs, such as the heart, kidney, and brain, which is inextricably related to calcium dyshomeostasis and downstream pathological events. Due to the increasing global burden of kidney or cardiovascular diseases (CVDs), safe and efficient drugs specific to novel targets are imperatively needed. Notably, investigation of the possibility to regard the TRPM2 channel as a new therapeutic target in ROS-related CVDs or renal diseases is urgently required because the roles of the TRPM2 channel in heart or kidney diseases have not received enough attention and thus have not been fully elaborated. Therefore, we aimed to review the involvement of the TRPM2 channel in cardiovascular disorders related to kidney or typical renal diseases and attempted to speculate about TRPM2-mediated mechanisms of cardiorenal syndrome (CRS) to provide representative perspectives for future research about novel and effective therapeutic strategies.
Collapse
Affiliation(s)
- Zihan Chen
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, China
| | - Zaihua Cheng
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Congcong Ding
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tianyu Cao
- Biological anthropology, University of California, Santa Barbara, CA, USA
| | - Ling Chen
- Department of Cardiology, the First People's Hospital of Jiujiang, Jiujiang, China
| | - Hong Wang
- Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Junpei Li
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Xiao Huang
- Department of Cardiology, the Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
14
|
Zernov N, Popugaeva E. Role of Neuronal TRPC6 Channels in Synapse Development, Memory Formation and Animal Behavior. Int J Mol Sci 2023; 24:15415. [PMID: 37895105 PMCID: PMC10607207 DOI: 10.3390/ijms242015415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The transient receptor potential cation channel, subfamily C, member 6 (TRPC6), has been believed to adjust the formation of an excitatory synapse. The positive regulation of TRPC6 engenders synapse enlargement and improved learning and memory in animal models. TRPC6 is involved in different synaptoprotective signaling pathways, including antagonism of N-methyl-D-aspartate receptor (NMDAR), activation of brain-derived neurotrophic factor (BDNF) and postsynaptic store-operated calcium entry. Positive regulation of TRPC6 channels has been repeatedly shown to be good for memory formation and storage. TRPC6 is mainly expressed in the hippocampus, particularly in the dentate granule cells, cornu Ammonis 3 (CA3) pyramidal cells and gamma-aminobutyric acid (GABA)ergic interneurons. It has been observed that TRPC6 agonists have a great influence on animal behavior including memory formation and storage The purpose of this review is to collect the available information on the role of TRPC6 in memory formation in various parts of the brain to understand how TRPC6-specific pharmaceutical agents will affect memory in distinct parts of the central nervous system (CNS).
Collapse
Affiliation(s)
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
15
|
Okada Y, Numata T, Sabirov RZ, Kashio M, Merzlyak PG, Sato-Numata K. Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 3: the roles and properties of TRPM2 and TRPM7. Front Cell Dev Biol 2023; 11:1246955. [PMID: 37842082 PMCID: PMC10576435 DOI: 10.3389/fcell.2023.1246955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell volume regulation (CVR) is a prerequisite for animal cells to survive and fulfill their functions. CVR dysfunction is essentially involved in the induction of cell death. In fact, sustained normotonic cell swelling and shrinkage are associated with necrosis and apoptosis, and thus called the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. Since a number of ubiquitously expressed ion channels are involved in the CVR processes, these volume-regulatory ion channels are also implicated in the NVI and AVD events. In Part 1 and Part 2 of this series of review articles, we described the roles of swelling-activated anion channels called VSOR or VRAC and acid-activated anion channels called ASOR or PAC in CVR and cell death processes. Here, Part 3 focuses on therein roles of Ca2+-permeable non-selective TRPM2 and TRPM7 cation channels activated by stress. First, we summarize their phenotypic properties and molecular structure. Second, we describe their roles in CVR. Since cell death induction is tightly coupled to dysfunction of CVR, third, we focus on their participation in the induction of or protection against cell death under oxidative, acidotoxic, excitotoxic, and ischemic conditions. In this regard, we pay attention to the sensitivity of TRPM2 and TRPM7 to a variety of stress as well as to their capability to physicall and functionally interact with other volume-related channels and membrane enzymes. Also, we summarize a large number of reports hitherto published in which TRPM2 and TRPM7 channels are shown to be involved in cell death associated with a variety of diseases or disorders, in some cases as double-edged swords. Lastly, we attempt to describe how TRPM2 and TRPM7 are organized in the ionic mechanisms leading to cell death induction and protection.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| | - Ravshan Z. Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Makiko Kashio
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
| | - Peter G. Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| |
Collapse
|
16
|
Foster HM, Carle MN, Jira LR, Koh DW. TRPM2 Channels: A Potential Therapeutic Target in Melanoma? Int J Mol Sci 2023; 24:10437. [PMID: 37445615 DOI: 10.3390/ijms241310437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
The transient receptor potential, the melastatin (TRPM) subfamily, which consists of eight known members, appears to have significant importance in melanoma progression, treatment, and prognosis. As several members were originally cloned from cancerous tissue, initial studies aimed towards identifying TRPM involvement in cancer progression and tumorigenesis. For relevance in skin cancer, previous research has shown roles for several TRPM members in skin cancer progression, growth, and patient prognosis. One unique member, TRPM2, appears to have notable therapeutic potential in the treatment of melanoma. Previous and recent studies have demonstrated increased TRPM2 expression levels in melanoma, as well as important roles for TRPM2 in melanoma growth, proliferation, and survival. TRPM2 is thus an emerging target in the treatment of melanoma, where TRPM2 antagonism may offer an additional treatment option for melanoma patients in the future.
Collapse
Affiliation(s)
- Hattie M Foster
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - McKenzie N Carle
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Lukas R Jira
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - David W Koh
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| |
Collapse
|
17
|
Morishita K, Watanabe K, Naguro I, Ichijo H. Sodium ion influx regulates liquidity of biomolecular condensates in hyperosmotic stress response. Cell Rep 2023; 42:112315. [PMID: 37019112 DOI: 10.1016/j.celrep.2023.112315] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/17/2023] [Accepted: 03/14/2023] [Indexed: 04/07/2023] Open
Abstract
Biomolecular condensates are membraneless structures formed through phase separation. Recent studies have demonstrated that the material properties of biomolecular condensates are crucial for their biological functions and pathogenicity. However, the phase maintenance of biomolecular condensates in cells remains elusive. Here, we show that sodium ion (Na+) influx regulates the condensate liquidity under hyperosmotic stress. ASK3 condensates have higher fluidity at the high intracellular Na+ concentration derived from extracellular hyperosmotic solution. Moreover, we identified TRPM4 as a cation channel that allows Na+ influx under hyperosmotic stress. TRPM4 inhibition causes the liquid-to-solid phase transition of ASK3 condensates, leading to impairment of the ASK3 osmoresponse. In addition to ASK3 condensates, intracellular Na+ widely regulates the condensate liquidity and aggregate formation of biomolecules, including DCP1A, TAZ, and polyQ-protein, under hyperosmotic stress. Our findings demonstrate that changes in Na+ contribute to the cellular stress response via liquidity maintenance of biomolecular condensates.
Collapse
Affiliation(s)
- Kazuhiro Morishita
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
18
|
Zhang Y, Han Y, Wang L, Kong J, Pan W, Zhang X, Chen L, Yao Z, Zhou T, Cao J. Flufenamic Acid, a Promising Agent for the Sensitization of Colistin-Resistant Gram-Negative Bacteria to Colistin. Microbiol Spectr 2023; 11:e0405222. [PMID: 36971552 PMCID: PMC10100705 DOI: 10.1128/spectrum.04052-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
The continuous development of multidrug-resistant (MDR) Gram-negative bacteria poses a serious risk to public health on a worldwide scale. Colistin is used as the last-line antibiotic for the treatment of MDR pathogens, and colistin-resistant (COL-R) bacterial emergence thus has the potential to have a severe adverse impact on patient outcomes. In this study, synergistic activity was observed when colistin and flufenamic acid (FFA) were combined and used for the in vitro treatment of clinical COL-R Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii strains, as shown by checkerboard and time-kill assays. Crystal violet staining and scanning electron microscopy revealed the synergistic action of colistin-FFA against biofilms. When used to treat murine RAW264.7 macrophages, this combination did not induce any adverse toxicity. Strikingly, the survival rates of bacterially infected Galleria mellonella larvae were improved by such combination treatment, which was also sufficient to reduce the measured bacterial loads in a murine thigh infection model. Mechanistic propidium iodide (PI) staining analysis further demonstrated the ability of these agents to alter bacterial permeability in a manner that enhanced the efficacy of colistin treatment. Together, these data thus demonstrate that colistin and FFA can be synergistically combined to combat the spread of COL-R Gram-negative bacteria, providing a promising therapeutic tool with the potential to protect against COL-R bacterial infections and improve patient outcomes. IMPORTANCE Colistin is a last-line antibiotic used for the treatment of MDR Gram-negative bacterial infections. However, increasing resistance to it has been observed during clinical treatment. In this work, we assessed the efficacy of the combination of colistin and FFA for the treatment of COL-R bacterial isolates, demonstrating that the combined treatment has effective antibacterial and antibiofilm activities. Due to its low cytotoxicity and good therapeutic effects in vitro, the colistin-FFA combination may be a potential candidate for research into a resistance-modifying agent to combat infections caused by COL-R Gram-negative bacteria.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yijia Han
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Lingbo Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Jingchun Kong
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wei Pan
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Xiaodong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Zhuocheng Yao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Jianming Cao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
19
|
Grohs L, Cheng L, Cönen S, Haddad BG, Bülow A, Toklucu I, Ernst L, Körner J, Schmalzing G, Lampert A, Machtens JP, Hausmann R. Diclofenac and other non-steroidal anti-inflammatory drugs (NSAIDs) are competitive antagonists of the human P2X3 receptor. Front Pharmacol 2023; 14:1120360. [PMID: 37007008 PMCID: PMC10060569 DOI: 10.3389/fphar.2023.1120360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction: The P2X3 receptor (P2X3R), an ATP-gated non-selective cation channel of the P2X receptor family, is expressed in sensory neurons and involved in nociception. P2X3R inhibition was shown to reduce chronic and neuropathic pain. In a previous screening of 2000 approved drugs, natural products, and bioactive substances, various non-steroidal anti-inflammatory drugs (NSAIDs) were found to inhibit P2X3R-mediated currents.Methods: To investigate whether the inhibition of P2X receptors contributes to the analgesic effect of NSAIDs, we characterized the potency and selectivity of various NSAIDs at P2X3R and other P2XR subtypes using two-electrode voltage clamp electrophysiology.Results: We identified diclofenac as a hP2X3R and hP2X2/3R antagonist with micromolar potency (with IC50 values of 138.2 and 76.7 µM, respectively). A weaker inhibition of hP2X1R, hP2X4R, and hP2X7R by diclofenac was determined. Flufenamic acid (FFA) inhibited hP2X3R, rP2X3R, and hP2X7R (IC50 values of 221 µM, 264.1 µM, and ∼900 µM, respectively), calling into question its use as a non-selective ion channel blocker, when P2XR-mediated currents are under study. Inhibition of hP2X3R or hP2X2/3R by diclofenac could be overcome by prolonged ATP application or increasing concentrations of the agonist α,β-meATP, respectively, indicating competition of diclofenac and the agonists. Molecular dynamics simulation showed that diclofenac largely overlaps with ATP bound to the open state of the hP2X3R. Our results suggest a competitive antagonism through which diclofenac, by interacting with residues of the ATP-binding site, left flipper, and dorsal fin domains, inhibits the gating of P2X3R by conformational fixation of the left flipper and dorsal fin domains. In summary, we demonstrate the inhibition of the human P2X3 receptor by various NSAIDs. Diclofenac proved to be the most effective antagonist with a strong inhibition of hP2X3R and hP2X2/3R and a weaker inhibition of hP2X1R, hP2X4R, and hP2X7R.Discussion: Considering their involvement in nociception, inhibition of hP2X3R and hP2X2/3R by micromolar concentrations of diclofenac, which are rarely reached in the therapeutic range, may play a minor role in analgesia compared to the high-potency cyclooxygenase inhibition but may explain the known side effect of taste disturbances caused by diclofenac.
Collapse
Affiliation(s)
- Laura Grohs
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Linhan Cheng
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
| | - Saskia Cönen
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
- Molecular and Cellular Physiology (IBI-1), Institute of Biological Information Processing (IBI), Forschungszentrum Jülich, Jülich, Germany
| | - Bassam G. Haddad
- Molecular and Cellular Physiology (IBI-1), Institute of Biological Information Processing (IBI), Forschungszentrum Jülich, Jülich, Germany
| | - Astrid Bülow
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
- Department of Plastic Surgery, Hand Surgery—Burn Center, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Idil Toklucu
- Institute of Physiology (Neurophysiology), RWTH Aachen University, Aachen, Germany
| | - Lisa Ernst
- Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University, Aachen, Germany
| | - Jannis Körner
- Institute of Physiology (Neurophysiology), RWTH Aachen University, Aachen, Germany
- Department of Anesthesiology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Günther Schmalzing
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology (Neurophysiology), RWTH Aachen University, Aachen, Germany
| | - Jan-Philipp Machtens
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
- Molecular and Cellular Physiology (IBI-1), Institute of Biological Information Processing (IBI), Forschungszentrum Jülich, Jülich, Germany
| | - Ralf Hausmann
- Institute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
- *Correspondence: Ralf Hausmann,
| |
Collapse
|
20
|
Waterloo L, Hübner H, Fierro F, Pfeiffer T, Brox R, Löber S, Weikert D, Niv MY, Gmeiner P. Discovery of 2-Aminopyrimidines as Potent Agonists for the Bitter Taste Receptor TAS2R14. J Med Chem 2023; 66:3499-3521. [PMID: 36847646 DOI: 10.1021/acs.jmedchem.2c01997] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The bitter taste receptor TAS2R14 is a G protein-coupled receptor that is found on the tongue as well as in the human airway smooth muscle and other extraoral tissues. Because its activation causes bronchodilatation, TAS2R14 is a potential target for the treatment of asthma or chronic obstructive pulmonary disease. Structural variations of flufenamic acid, a nonsteroidal anti-inflammatory drug, led us to 2-aminopyridines showing considerable efficacy and potency in an IP1accumulation assay. In combination with an exchange of the carboxylic moiety by a tetrazole unit, a set of promising new TAS2R14 agonists was developed. The most potent ligand 28.1 (EC50 = 72 nM) revealed a six-fold higher potency than flufenamic acid and a maximum efficacy of 129%. Besides its unprecedented TAS2R14 activation, 28.1 revealed marked selectivity over a panel of 24 non-bitter taste human G protein-coupled receptors.
Collapse
Affiliation(s)
- Lukas Waterloo
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| | - Fabrizio Fierro
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry, Food Science and Nutrition, The Hebrew University, Rehovot 7610001, Israel
| | - Tara Pfeiffer
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| | - Regine Brox
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| | - Stefan Löber
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| | - Masha Y Niv
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry, Food Science and Nutrition, The Hebrew University, Rehovot 7610001, Israel
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, Erlangen 91058, Germany
| |
Collapse
|
21
|
TRPC absence induces pro-inflammatory macrophages and gut microbe disorder, sensitizing mice to colitis. Int Immunopharmacol 2023; 115:109655. [PMID: 36592529 DOI: 10.1016/j.intimp.2022.109655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023]
Abstract
The transient receptor potential canonical (TRPC) channels, encoded in seven non-allelic genes, are important contributors to calcium fluxes, are strongly associated with various diseases. Here we explored the consequences of ablating all seven TRPCs in mice focusing on colitis. We discovered that absence of all seven TRPC proteins in mice (TRPC HeptaKO mice) promotes the development of dextran sulfate sodium (DSS)-induced colitis. RNA-sequence analysis highlighted an extremely pro-inflammatory profile in colons of DSS-treated TRPC HeptaKO mice, with an amount of increased pro-inflammatory cytokines and chemokines. Flow cytometry analysis showed that the infiltration of Ly6Chi monocytes and neutrophils in colonic lamina propria was significantly increased in DSS-treated TRPC HeptaKO mice. Results also revealed that macrophages from TRPC HeptaKO mice exhibited M1 polarization and enhanced secretion of pro-inflammatory factors. In addition, the composition of gut microbiota was markedly disturbed in DSS-treated TRPC HeptaKO mice. However, upon antibiotic cocktail (Abx)-treatment, TRPC HeptaKO mice showed no significant differences with WT mice in disease severity. Collectively, these data suggest that ablation of all TRPCs promotes the development of DSS-induced colitis by inducing pro-inflammatory macrophages and gut microbiota disorder.
Collapse
|
22
|
Snyder RR, Blitz DM. Multiple intrinsic membrane properties are modulated in a switch from single- to dual-network activity. J Neurophysiol 2022; 128:1181-1198. [PMID: 36197020 PMCID: PMC9621714 DOI: 10.1152/jn.00337.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/14/2022] [Accepted: 10/01/2022] [Indexed: 11/22/2022] Open
Abstract
Neural network flexibility includes changes in neuronal participation between networks, such as the switching of neurons between single- and dual-network activity. We previously identified a neuron that is recruited to burst in time with an additional network via modulation of its intrinsic membrane properties, instead of being recruited synaptically into the second network. However, the modulated intrinsic properties were not determined. Here, we use small networks in the Jonah crab (Cancer borealis) stomatogastric nervous system (STNS) to examine modulation of intrinsic properties underlying neuropeptide (Gly1-SIFamide)-elicited neuronal switching. The lateral posterior gastric neuron (LPG) switches from exclusive participation in the fast pyloric (∼1 Hz) network, due to electrical coupling, to dual-network activity that includes periodic escapes from the fast rhythm via intrinsically generated oscillations at the slower gastric mill network frequency (∼0.1 Hz). We isolated LPG from both networks by pharmacology and hyperpolarizing current injection. Gly1-SIFamide increased LPG intrinsic excitability and rebound from inhibition and decreased spike frequency adaptation, which can all contribute to intrinsic bursting. Using ion substitution and channel blockers, we found that a hyperpolarization-activated current, a persistent sodium current, and calcium or calcium-related current(s) appear to be primary contributors to Gly1-SIFamide-elicited LPG intrinsic bursting. However, this intrinsic bursting was more sensitive to blocking currents when LPG received rhythmic electrical coupling input from the fast network than in the isolated condition. Overall, a switch from single- to dual-network activity can involve modulation of multiple intrinsic properties, while synaptic input from a second network can shape the contributions of these properties.NEW & NOTEWORTHY Neuropeptide-elicited intrinsic bursting was recently determined to switch a neuron from single- to dual-network participation. Here we identified multiple intrinsic properties modulated in the dual-network state and candidate ion channels underlying the intrinsic bursting. Bursting at the second network frequency was more sensitive to blocking currents in the dual-network state than when neurons were synaptically isolated from their home network. Thus, synaptic input can shape the contributions of modulated intrinsic properties underlying dual-network activity.
Collapse
Affiliation(s)
- Ryan R Snyder
- Department of Biology and Center for Neuroscience, Miami University, Oxford, Ohio
| | - Dawn M Blitz
- Department of Biology and Center for Neuroscience, Miami University, Oxford, Ohio
| |
Collapse
|
23
|
Chen J, Chang Y, Zhu J, Peng Y, Li Z, Zhang K, Zhang Y, Lin C, Lin Z, Pan S, Huang K. Flufenamic acid improves survival and neurologic outcome after successful cardiopulmonary resuscitation in mice. J Neuroinflammation 2022; 19:214. [PMID: 36050694 PMCID: PMC9438280 DOI: 10.1186/s12974-022-02571-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background Brain injury is the main cause of high mortality and disability after successful cardiopulmonary resuscitation (CPR) from sudden cardiac arrest (CA). The transient receptor potential M4 (TRPM4) channel is a novel target for ameliorating blood–brain barrier (BBB) disruption and neuroinflammation. Herein, we tested whether flufenamic acid (FFA), which is reported to block TRPM4 with high potency, could confer neuroprotection against brain injury secondary to CA/CPR and whether its action was exerted by blocking the TRPM4 channel. Methods Wild-type (WT) and Trpm4 knockout (Trpm4−/−) mice subjected to 10-min CA/CPR were randomized to receive FFA or vehicle once daily. Post-CA/CPR brain injuries including neurologic deficits, survival rate, histological damage, edema formation, BBB destabilization and neuroinflammation were assessed. Results In WT mice subjected to CA/CPR, FFA was effective in improving survival and neurologic outcome, reducing neuropathological injuries, attenuating brain edema, lessening the leakage of IgG and Evans blue dye, restoring tight junction protein expression and promoting microglia/macrophages from the pro-inflammatory subtype toward the anti-inflammatory subtype. In comparison to WT mice, Trpm4−/− mice exhibited less neurologic deficiency, milder histological impairment, more BBB integrity and more anti-inflammatory microglia/macrophage polarization. As expected, FFA did not provide a benefit of superposition compared with vehicle in the Trpm4−/− mice after CA/CPR. Conclusions FFA mitigates BBB breach and modifies the functional status of microglia/macrophages, thereby improving survival and neurologic deficits following CA/CPR. The neuroprotective effects occur at least partially by interfering with the TRPM4 channel in the neurovascular unit. These results indicate the significant clinical potential of FFA to improve the prognosis for CA victims who are successfully resuscitated. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02571-2.
Collapse
Affiliation(s)
- Jiancong Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Juan Zhu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuqin Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Zheqi Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Yuzhen Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Chuman Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Zhenzhou Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China.
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou North Avenue, Guangzhou, 1838#510515, China.
| |
Collapse
|
24
|
Barioni NO, Derakhshan F, Tenorio Lopes L, Onimaru H, Roy A, McDonald F, Scheibli E, Baghdadwala MI, Heidari N, Bharadia M, Ikeda K, Yazawa I, Okada Y, Harris MB, Dutschmann M, Wilson RJA. Novel oxygen sensing mechanism in the spinal cord involved in cardiorespiratory responses to hypoxia. SCIENCE ADVANCES 2022; 8:eabm1444. [PMID: 35333571 PMCID: PMC8956269 DOI: 10.1126/sciadv.abm1444] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 02/04/2022] [Indexed: 05/05/2023]
Abstract
As blood oxygenation decreases (hypoxemia), mammals mount cardiorespiratory responses, increasing oxygen to vital organs. The carotid bodies are the primary oxygen chemoreceptors for breathing, but sympathetic-mediated cardiovascular responses to hypoxia persist in their absence, suggesting additional high-fidelity oxygen sensors. We show that spinal thoracic sympathetic preganglionic neurons are excited by hypoxia and silenced by hyperoxia, independent of surrounding astrocytes. These spinal oxygen sensors (SOS) enhance sympatho-respiratory activity induced by CNS asphyxia-like stimuli, suggesting they bestow a life-or-death advantage. Our data suggest the SOS use a mechanism involving neuronal nitric oxide synthase 1 (NOS1) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). We propose NOS1 serves as an oxygen-dependent sink for NADPH in hyperoxia. In hypoxia, NADPH catabolism by NOS1 decreases, increasing availability of NADPH to NOX and launching reactive oxygen species-dependent processes, including transient receptor potential channel activation. Equipped with this mechanism, SOS are likely broadly important for physiological regulation in chronic disease, spinal cord injury, and cardiorespiratory crisis.
Collapse
Affiliation(s)
- Nicole O. Barioni
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Fatemeh Derakhshan
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Luana Tenorio Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Arijit Roy
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Fiona McDonald
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Erika Scheibli
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mufaddal I. Baghdadwala
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Negar Heidari
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Manisha Bharadia
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keiko Ikeda
- Division of Internal Medicine, Murayama Medical Center, Musashimurayama, Tokyo, Japan
| | - Itaru Yazawa
- Global Research Center for Innovative Life Science, Peptide Drug Innovation, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo 142-8501, Japan
| | - Yasumasa Okada
- Division of Internal Medicine, Murayama Medical Center, Musashimurayama, Tokyo, Japan
| | - Michael B. Harris
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Mathias Dutschmann
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Richard J. A. Wilson
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
25
|
Hyperthermia and Serotonin: The Quest for a “Better Cyproheptadine”. Int J Mol Sci 2022; 23:ijms23063365. [PMID: 35328784 PMCID: PMC8952796 DOI: 10.3390/ijms23063365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Fine temperature control is essential in homeothermic animals. Both hyper- and hypothermia can have deleterious effects. Multiple, efficient and partly redundant mechanisms of adjusting the body temperature to the value set by the internal thermostat exist. The neural circuitry of temperature control and the neurotransmitters involved are reviewed. The GABAergic inhibitory output from the brain thermostat in the preoptic area POA to subaltern neural circuitry of temperature control (Nucleus Raphe Dorsalis and Nucleus Raphe Pallidus) is a function of the balance between the (opposite) effects mediated by the transient receptor potential receptor TRPM2 and EP3 prostaglandin receptors. Activation of TRPM2-expressing neurons in POA favors hypothermia, while inhibition has the opposite effect. Conversely, EP3 receptors induce elevation in body temperature. Activation of EP3-expressing neurons in POA results in hyperthermia, while inhibition has the opposite effect. Agonists at TRPM2 and/or antagonists at EP3 could be beneficial in hyperthermia control. Activity of the neural circuitry of temperature control is modulated by a variety of 5-HT receptors. Based on the theoretical model presented the “ideal” antidote against serotonin syndrome hyperthermia appears to be an antagonist at the 5-HT receptor subtypes 2, 4 and 6 and an agonist at the receptor subtypes 1, 3 and 7. Very broadly speaking, such a profile translates in a sympatholytic effect. While a compound with such an ideal profile is presently not available, better matches than the conventional antidote cyproheptadine (used off-label in severe serotonin syndrome cases) appear to be possible and need to be identified.
Collapse
|
26
|
Maestrelli F, Cirri M, De Luca E, Biagi D, Mura P. Role of Cyclodextrins and Drug Solid State Properties on Flufenamic Acid Dissolution Performance from Tablets. Pharmaceutics 2022; 14:pharmaceutics14020284. [PMID: 35214017 PMCID: PMC8880332 DOI: 10.3390/pharmaceutics14020284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
Flufenamic acid (FFA) is a non-steroidal anti-inflammatory drug characterised by a low solubility and problems of variable dissolution rate and bio-inequivalence. Different FFA batches, obtained by different suppliers, showed different powder characteristics (particle size, shape and surface properties) that may affect its dissolution behaviour from solid dosage forms. Aim of this work was the improvement of FFA solubility and dissolution rate by the use of cyclodextrins (CDs) and the obtainment of an effective tablet formulation by direct compression. Several CDs have been tested, both in solution and in solid state and several binary systems drug-CDs have been obtained with different techniques, with the scope to select the most effective system. Grinding technique with randomly methylated-β-cyclodextrin (RAMEB) was the only one that allowed the complete drug amorphization, together with the highest improvement in drug dissolution rate, and was then selected for tablets formulation. Conventional and immediate release tablets were obtained and fully characterised for technological properties. In both cases an improved and well reproducible drug dissolution performance was obtained, independently from the FFA supplier and thus no more affected by the differences observed between the original FFA crystalline samples.
Collapse
Affiliation(s)
- Francesca Maestrelli
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
| | - Marzia Cirri
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
- Correspondence:
| | - Enrico De Luca
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
| | - Diletta Biagi
- Menarini Manufacturing Logistic and Services s.r.l. (AMMLS), Via dei Sette Santi 1/3, 50131 Florence, Italy;
| | - Paola Mura
- Department of Chemistry “U. Schiff”, University of Florence, Via Schiff 6, Sesto Fiorentino, 50019 Florence, Italy; (F.M.); (E.D.L.); (P.M.)
| |
Collapse
|
27
|
Kovács ZM, Dienes C, Hézső T, Almássy J, Magyar J, Bányász T, Nánási PP, Horváth B, Szentandrássy N. Pharmacological Modulation and (Patho)Physiological Roles of TRPM4 Channel—Part 1: Modulation of TRPM4. Pharmaceuticals (Basel) 2022; 15:ph15010081. [PMID: 35056138 PMCID: PMC8781449 DOI: 10.3390/ph15010081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential melastatin 4 is a unique member of the TRPM protein family and, similarly to TRPM5, is Ca2+-sensitive and permeable to monovalent but not divalent cations. It is widely expressed in many organs and is involved in several functions by regulating the membrane potential and Ca2+ homeostasis in both excitable and non-excitable cells. This part of the review discusses the pharmacological modulation of TRPM4 by listing, comparing, and describing both endogenous and exogenous activators and inhibitors of the ion channel. Moreover, other strategies used to study TRPM4 functions are listed and described. These strategies include siRNA-mediated silencing of TRPM4, dominant-negative TRPM4 variants, and anti-TRPM4 antibodies. TRPM4 is receiving more and more attention and is likely to be the topic of research in the future.
Collapse
Affiliation(s)
- Zsigmond Máté Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.M.K.); (C.D.); (T.H.); (J.A.); (J.M.); (T.B.); (P.P.N.); (B.H.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence:
| |
Collapse
|
28
|
Hernandez A, Alaniz-Palacios A, Contreras-Vite JA, Martínez-Torres A. Positive modulation of the TMEM16B mediated currents by TRPV4 antagonist. Biochem Biophys Rep 2021; 28:101180. [PMID: 34917777 PMCID: PMC8646129 DOI: 10.1016/j.bbrep.2021.101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
Calcium-activated chloride channels (CaCCs) play important roles in many physiological processes and their malfunction is implicated in diverse pathologies such as cancer, asthma, and hypertension. TMEM16A and TMEM16B proteins are the structural components of the CaCCs. Recent studies in cell cultures and animal models have demonstrated that pharmacological inhibition of CaCCs could be helpful in the treatment of some diseases, however, there are few specific modulators of these channels. CaCCs and Transient Receptor Potential Vanilloid-4 (TRPV4) channels are co-expressed in some tissues where they functionally interact. TRPV4 is activated by different stimuli and forms a calcium permeable channel that is activated by GSK1016790A and antagonized by GSK2193874. Here we report that GSK2193874 enhances the chloride currents mediated by TMEM16B expressed in HEK cells at nanomolar concentrations and that GSK1016790A enhances native CaCCs of Xenopus oocytes. Thus, these compounds may be used as a tool for the study of CaCCs, TRPV4 and their interactions.
Collapse
Affiliation(s)
- Adan Hernandez
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, 76230 Santiago de Querétaro, Querétaro, Mexico
| | - Alfredo Alaniz-Palacios
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, 76230 Santiago de Querétaro, Querétaro, Mexico
| | - Juan A Contreras-Vite
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, 76230 Santiago de Querétaro, Querétaro, Mexico
| | - Ataúlfo Martínez-Torres
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, 76230 Santiago de Querétaro, Querétaro, Mexico
| |
Collapse
|
29
|
Wang Q, Liu N, Ni YS, Yang JM, Ma L, Lan XB, Wu J, Niu JG, Yu JQ. TRPM2 in ischemic stroke: Structure, molecular mechanisms, and drug intervention. Channels (Austin) 2021; 15:136-154. [PMID: 33455532 PMCID: PMC7833771 DOI: 10.1080/19336950.2020.1870088] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke has a high lethality rate worldwide, and novel treatments are limited. Calcium overload is considered to be one of the mechanisms of cerebral ischemia. Transient receptor potential melastatin 2 (TRPM2) is a reactive oxygen species (ROS)-sensitive calcium channel. Cerebral ischemia-induced TRPM2 activation triggers abnormal intracellular Ca2+ accumulation and cell death, which in turn causes irreversible brain damage. Thus, TRPM2 has emerged as a new therapeutic target for ischemic stroke. This review provides data on the expression, structure, and function of TRPM2 and illustrates its cellular and molecular mechanisms in ischemic stroke. Natural and synthetic TRPM2 inhibitors (both specific and nonspecific) are also summarized. The three-dimensional protein structure of TRPM2 has been identified, and we speculate that molecular simulation techniques will be essential for developing new drugs that block TRPM2 channels. These insights about TRPM2 may be the key to find potent therapeutic approaches for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qing Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Yuan-Shu Ni
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jia-Mei Yang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Lin Ma
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Xiao-Bing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jing Wu
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, China
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Jian-Qiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
- Ningxia Collaborative Innovation Center of Regional Characteristic Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
30
|
Nardin C, Peres C, Putti S, Orsini T, Colussi C, Mazzarda F, Raspa M, Scavizzi F, Salvatore AM, Chiani F, Tettey-Matey A, Kuang Y, Yang G, Retamal MA, Mammano F. Connexin Hemichannel Activation by S-Nitrosoglutathione Synergizes Strongly with Photodynamic Therapy Potentiating Anti-Tumor Bystander Killing. Cancers (Basel) 2021; 13:cancers13205062. [PMID: 34680212 PMCID: PMC8533914 DOI: 10.3390/cancers13205062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Bystander effects depend on direct cell-cell communication and/or paracrine signaling mediated by the release of soluble factors into the extracellular environment and may greatly influence therapy outcome. Although the limited data available suggest a role for intercellular gap junction channels, far less is known about the role of connexin hemichannels. Here, we investigated bystander effects induced by photodynamic therapy in syngeneic murine melanoma models in vivo. We determined that (i) photoactivation of a photosensitizer triggered calcium-dependent cell death pathways in both irradiated and bystander tumor cells; (ii) hemichannel activity and adenosine triphosphate release were key factors for the induction of bystander cell death; and (iii) bystander cell killing and antitumor response elicited by photodynamic therapy were greatly enhanced by combination treatment with S-nitrosoglutathione, which promoted hemichannel opening in these experimental conditions. Therefore, these findings in a preclinical model have important translational potential. Abstract In this study, we used B16-F10 cells grown in the dorsal skinfold chamber (DSC) preparation that allowed us to gain optical access to the processes triggered by photodynamic therapy (PDT). Partial irradiation of a photosensitized melanoma triggered cell death in non-irradiated tumor cells. Multiphoton intravital microscopy with genetically encoded fluorescence indicators revealed that bystander cell death was mediated by paracrine signaling due to adenosine triphosphate (ATP) release from connexin (Cx) hemichannels (HCs). Intercellular calcium (Ca2+) waves propagated from irradiated to bystander cells promoting intracellular Ca2+ transfer from the endoplasmic reticulum (ER) to mitochondria and rapid activation of apoptotic pathways. Combination treatment with S-nitrosoglutathione (GSNO), an endogenous nitric oxide (NO) donor that biases HCs towards the open state, greatly potentiated anti-tumor bystander killing via enhanced Ca2+ signaling, leading to a significant reduction of post-irradiation tumor mass. Our results demonstrate that HCs can be exploited to dramatically increase cytotoxic bystander effects and reveal a previously unappreciated role for HCs in tumor eradication promoted by PDT.
Collapse
Affiliation(s)
- Chiara Nardin
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Chiara Peres
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Sabrina Putti
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Tiziana Orsini
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Claudia Colussi
- Institute for Systems Analysis and Computer Science “A. Ruberti” (IASI)-CNR, 00168 Rome, Italy;
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Flavia Mazzarda
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Anna Maria Salvatore
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Francesco Chiani
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Abraham Tettey-Matey
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
| | - Yuanyuan Kuang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; (Y.K.); (G.Y.)
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; (Y.K.); (G.Y.)
| | - Mauricio A. Retamal
- Universidad del Desarrollo, Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana, Santiago 7780272, Chile;
| | - Fabio Mammano
- Institute of Biochemistry and Cell Biology (IBBC)-CNR, 00015 Rome, Italy; (C.N.); (C.P.); (S.P.); (T.O.); (F.M.); (M.R.); (F.S.); (A.M.S.); (F.C.); (A.T.-M.)
- Department of Physics and Astronomy “G. Galilei”, University of Padova, 35131 Padova, Italy
- Correspondence:
| |
Collapse
|
31
|
Sui S, Yu H, Wang X, Wang W, Yang X, Pan X, Zhou Q, Xin C, Du R, Wu S, Zhang J, Cao Q, Wang N, Kuehn MH, Zhu W. iPSC-Derived Trabecular Meshwork Cells Stimulate Endogenous TM Cell Division Through Gap Junction in a Mouse Model of Glaucoma. Invest Ophthalmol Vis Sci 2021; 62:28. [PMID: 34427623 PMCID: PMC8399400 DOI: 10.1167/iovs.62.10.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/28/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose Decreased trabecular meshwork (TM) cellularity has been implicated as a major reason for TM dysfunction and aqueous humor (AH) outflow abnormalities in primary open angle glaucoma. We previously found that transplantation of induced pluripotent stem cell (iPSC)-derived TM cells can restore TM function and stimulate endogenous TM cell division. The goal of the present study is to investigate whether signaling via gap junctions is involved in this process. Methods Differentiated iPSCs were characterized morphologically, transcriptionally, and immunohistochemically. After purification, iPSC-TM were co-cultured with mouse TM (MTM) cells to mimic the transplantation procedure. Through the pharmacological antagonists and short hairpin RNA (shRNA) technique, the gap junction function in iPSC-based therapy was determined. Results In the co-culture system, iPSC-TM increase MTM cell division as well as transfer of Ca2+ to MTM. This effect was blocked by treatment with the gap junction inhibitors carbenoxolone (CBX) or flufenamic acid (FFA). The shRNA mediated knock down of connexin 43 (Cx43) expression in iPSC-TM also results in decreased Ca2+ transfer and lower MTM proliferation rates. In vivo, Cx43 downregulation in transplanted iPSC-TM weakened their regenerative role in an Ad5.myocilinY437H mouse model of glaucoma. Mice receiving these cells exhibited lower TM cellularity and higher intraocular pressure (IOP) than those receiving unmodified iPSC-TM. Conclusions Our findings reveal a crucial role of gap junction, especially Cx43, in iPSC-based TM regeneration, and provides insights to enhance the regenerative effect of iPSCs in glaucoma therapy.
Collapse
Affiliation(s)
- Shangru Sui
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Hongxia Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangji Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Wenyan Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Xuejiao Yang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaojing Pan
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Qingjun Zhou
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Chen Xin
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing, China
| | - Rong Du
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing, China
| | - Jingxue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing, China
| | - Qilong Cao
- Qingdao Haier Biotech Co. Ltd., Qingdao, China
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing, China
| | - Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Wei Zhu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beijing University & Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Pinto MC, Silva IAL, Figueira MF, Amaral MD, Lopes-Pacheco M. Pharmacological Modulation of Ion Channels for the Treatment of Cystic Fibrosis. J Exp Pharmacol 2021; 13:693-723. [PMID: 34326672 PMCID: PMC8316759 DOI: 10.2147/jep.s255377] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-shortening monogenic disease caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein, an anion channel that transports chloride and bicarbonate across epithelia. Despite clinical progress in delaying disease progression with symptomatic therapies, these individuals still develop various chronic complications in lungs and other organs, which significantly restricts their life expectancy and quality of life. The development of high-throughput assays to screen drug-like compound libraries have enabled the discovery of highly effective CFTR modulator therapies. These novel therapies target the primary defect underlying CF and are now approved for clinical use for individuals with specific CF genotypes. However, the clinically approved modulators only partially reverse CFTR dysfunction and there is still a considerable number of individuals with CF carrying rare CFTR mutations who remain without any effective CFTR modulator therapy. Accordingly, additional efforts have been pursued to identify novel and more potent CFTR modulators that may benefit a larger CF population. The use of ex vivo individual-derived specimens has also become a powerful tool to evaluate novel drugs and predict their effectiveness in a personalized medicine approach. In addition to CFTR modulators, pro-drugs aiming at modulating alternative ion channels/transporters are under development to compensate for the lack of CFTR function. These therapies may restore normal mucociliary clearance through a mutation-agnostic approach (ie, independent of CFTR mutation) and include inhibitors of the epithelial sodium channel (ENaC), modulators of the calcium-activated channel transmembrane 16A (TMEM16, or anoctamin 1) or of the solute carrier family 26A member 9 (SLC26A9), and anionophores. The present review focuses on recent progress and challenges for the development of ion channel/transporter-modulating drugs for the treatment of CF.
Collapse
Affiliation(s)
- Madalena C Pinto
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Iris A L Silva
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Miriam F Figueira
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Margarida D Amaral
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| |
Collapse
|
33
|
Shi R, Fu Y, Zhao D, Boczek T, Wang W, Guo F. Cell death modulation by transient receptor potential melastatin channels TRPM2 and TRPM7 and their underlying molecular mechanisms. Biochem Pharmacol 2021; 190:114664. [PMID: 34175300 DOI: 10.1016/j.bcp.2021.114664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Transient receptor potential melastatin (TRPM) channels are members of the transient receptor potential (TRP) channels, a family of evolutionarily conserved integral membrane proteins. TRPM channels are nonselective cation channels, mediating the influx of various ions including Ca2+, Na+ and Zn2+. The function of TRPM channels is vital for cell proliferation, cell development and cell death. Cell death is a key procedure during embryonic development, organism homeostasis, aging and disease. The category of cell death modalities, beyond the traditionally defined concepts of necrosis, autophagy, and apoptosis, were extended with the discovery of pyroptosis, necroptosis and ferroptosis. As upstream signaling regulators of cell death, TRPM channels have been involved inrelevant pathologies. In this review, we introduced several cell death modalities, then summarized the contribution of TRPM channels (especially TRPM2 and TRPM7) to different cell death modalities and discussed the underlying regulatory mechanisms. Our work highlighted the possibility of TRPM channels as potential therapeutic targets in cell death-related diseases.
Collapse
Affiliation(s)
- Ruixue Shi
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yu Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Dongyi Zhao
- The University of Tokyo, Department of Pharmaceutical Science, 1130033, Japan
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, 92215, Poland.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
34
|
Chappe Y, Michel P, Joushomme A, Barbeau S, Pierredon S, Baron L, Garenne A, Poulletier De Gannes F, Hurtier A, Mayer S, Lagroye I, Quignard JF, Ducret T, Compan V, Franchet C, Percherancier Y. High-throughput screening of TRPV1 ligands in the light of the Bioluminescence Resonance Energy Transfer technique. Mol Pharmacol 2021; 100:237-257. [PMID: 34127538 DOI: 10.1124/molpharm.121.000271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/17/2021] [Indexed: 11/22/2022] Open
Abstract
Ion channels are attractive drug targets for many therapeutic applications. However, high-throughput screening (HTS) of drug candidates is difficult and remains very expensive. We thus assessed the suitability of the Bioluminescence Resonance Energy Transfer (BRET) technique as a new HTS method for ion-channel studies by taking advantage of our recently characterized intra- and intermolecular BRET probes targeting the TRPV1 ion channel. These BRET probes monitor conformational changes during TRPV1 gating and subsequent coupling with Calmodulin, two molecular events that are intractable using reference techniques such as automated calcium assay (ACA) and automated patch-clamp (APC). We screened the small-sized Prestwick chemical library, encompassing 1200 compounds with high structural diversity, using either intra- and intermolecular BRET probes or ACA. Secondary screening of the detected hits was done using APC. Multiparametric analysis of our results shed light on the capability of calmodulin inhibitors included in the Prestwick library to inhibit TRPV1 activation by Capsaicin (CAPS). BRET was the lead technique for this identification process. Finally, we present data exemplifying the use of intramolecular BRET probes to study other TRPs and non-TRPs ion channels. Knowing the ease of use of BRET biosensors and the low cost of the BRET technique, these assays may advantageously be included for extending ion-channel drug screening. Significance Statement We screened a chemical library against TRPV1 ion channel using Bioluminescence Resonance Energy Transfer (BRET) molecular probes, and compared the results with the ones obtained using reference techniques such as automated calcium assay and automated patch-clamp. Multiparametric analysis of our results shed light on the capability of Calmodulin antagonists to inhibit chemical activation of TRPV1, and indicates that BRET probes may advantageously be included in ion channel drug screening campaigns.
Collapse
Affiliation(s)
- Yann Chappe
- IMS laboratory / CNRS UMR 5218, Bordeaux University, France
| | | | | | - Solène Barbeau
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, France
| | - Sandra Pierredon
- CNRS UMR 5203 - INSERM U1191, Institut de Genomique Fonctionnelle, France
| | | | - André Garenne
- IMS laboratory / CNRS UMR 5218, Bordeaux University, France
| | | | | | | | | | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Bordeaux University, France
| | - Vincent Compan
- CNRS UMR 5203 - INSERM U1191, Institut de Genomique Fonctionnelle, France
| | | | | |
Collapse
|
35
|
Barilli A, Aldegheri L, Bianchi F, Brault L, Brodbeck D, Castelletti L, Feriani A, Lingard I, Myers R, Nola S, Piccoli L, Pompilio D, Raveglia LF, Salvagno C, Tassini S, Virginio C, Sabat M. From High-Throughput Screening to Target Validation: Benzo[ d]isothiazoles as Potent and Selective Agonists of Human Transient Receptor Potential Cation Channel Subfamily M Member 5 Possessing In Vivo Gastrointestinal Prokinetic Activity in Rodents. J Med Chem 2021; 64:5931-5955. [PMID: 33890770 DOI: 10.1021/acs.jmedchem.1c00065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transient receptor potential cation channel subfamily M member 5 (TRPM5) is a nonselective monovalent cation channel activated by intracellular Ca2+ increase. Within the gastrointestinal system, TRPM5 is expressed in the stoma, small intestine, and colon. In the search for a selective agonist of TRPM5 possessing in vivo gastrointestinal prokinetic activity, a high-throughput screening was performed and compound 1 was identified as a promising hit. Hit validation and hit to lead activities led to the discovery of a series of benzo[d]isothiazole derivatives. Among these, compounds 61 and 64 showed nanomolar activity and excellent selectivity (>100-fold) versus related cation channels. The in vivo drug metabolism and pharmacokinetic profile of compound 64 was found to be ideal for a compound acting locally at the intestinal level, with minimal absorption into systemic circulation. Compound 64 was tested in vivo in a mouse motility assay at 100 mg/kg, and demonstrated increased prokinetic activity.
Collapse
Affiliation(s)
- Alessio Barilli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laura Aldegheri
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Federica Bianchi
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laurent Brault
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Daniela Brodbeck
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laura Castelletti
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Aldo Feriani
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Iain Lingard
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Richard Myers
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Selena Nola
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laura Piccoli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Daniela Pompilio
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Luca F Raveglia
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Cristian Salvagno
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Sabrina Tassini
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Caterina Virginio
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Mark Sabat
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| |
Collapse
|
36
|
Huang L, Xu G, Jiang R, Luo Y, Zuo Y, Liu J. Development of Non-opioid Analgesics Targeting Two-pore Domain Potassium Channels. Curr Neuropharmacol 2021; 20:16-26. [PMID: 33827408 PMCID: PMC9199554 DOI: 10.2174/1570159x19666210407152528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Two-pore domain potassium (K2P) channels are a diverse family of potassium channels. K2P channels generate background leak potassium currents to regulate cellular excitability and are thereby involved in a wide range of neurological disorders. K2P channels are modulated by a variety of physicochemical factors such as mechanical stretch, temperature, and pH. In the the peripheral nervous system (PNS), K2P channels are widely expressed in nociceptive neurons and play a critical roles in pain perception. In this review, we summarize the recent advances in the pharmacological properties of K2P channels, with a focus on the exogenous small-molecule activators targeting K2P channels. We emphasize the subtype-selectivity, cellular and in vivo pharmacological properties of all the reported small-molecule activators. The key underlying analgesic mechanisms mediated by K2P are also summarized based on the data in the literature from studies using small-molecule activators and genetic knock-out animals. We discuss advantages and limitations of the translational perspectives of K2P in pain medicine and provide outstanding questions for future studies in the end.
Collapse
Affiliation(s)
- Lu Huang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Guangyin Xu
- Department of Physiology and Neurobiology, Institute of Neuroscience, Medical College of Soochow University, Suzhou, 215123, Jiangsu. China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Yuncheng Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Yunxia Zuo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| |
Collapse
|
37
|
Abstract
Members of the transient receptor potential (TRP) channels that are expressed in the kidney have gained prominence in recent years following discoveries of their role in maintaining the integrity of the filtration barrier, regulating tubular reabsorption of Ca2+ and Mg2+, and sensing osmotic stimuli. Furthermore, evidence has linked mutations in TRP channels to kidney disease pathophysiological mechanisms, including focal segmental glomerulosclerosis, disturbances in Mg2+ homeostasis, and polycystic kidney disease. Several subtypes of TRP channels are expressed in the renal vasculature, from preglomerular arteries and arterioles to the descending vasa recta. Although investigations on the physiological and pathological significance of renal vascular TRP channels are sparse, studies on isolated vessels and cells have suggested their involvement in renal vasoregulation. Renal blood flow (RBF) is an essential determinant of kidney function, including glomerular filtration, water and solute reabsorption, and waste product excretion. Functional alterations in ion channels that are expressed in the endothelium and smooth muscle of renal vessels can modulate renal vascular resistance, arterial pressure, and RBF. Hence, renal vascular TRP channels are potential therapeutic targets for the treatment of kidney disease. This review summarizes the current knowledge of TRP channel expression in renal vasculature and their role in controlling kidney function in health and disease. TRP channels are widely distributed in mammalian kidneys in glomerular, tubular, and vascular cells. TRPC and TRPV channels are functionally expressed in afferent arterioles. TRPC4 may regulate Ca2+ signaling in the descending vasa recta. Smooth muscle, endothelial, and pericyte TRP channels may participate in signal transduction mechanisms. TRP channels underlie renal autoregulation and regional kidney perfusion in health and disease.
Collapse
Affiliation(s)
- Praghalathan Kanthakumar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Adebowale Adebiyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
38
|
Jimenez I, Prado Y, Marchant F, Otero C, Eltit F, Cabello-Verrugio C, Cerda O, Simon F. TRPM Channels in Human Diseases. Cells 2020; 9:E2604. [PMID: 33291725 PMCID: PMC7761947 DOI: 10.3390/cells9122604] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
The transient receptor potential melastatin (TRPM) subfamily belongs to the TRP cation channels family. Since the first cloning of TRPM1 in 1989, tremendous progress has been made in identifying novel members of the TRPM subfamily and their functions. The TRPM subfamily is composed of eight members consisting of four six-transmembrane domain subunits, resulting in homomeric or heteromeric channels. From a structural point of view, based on the homology sequence of the coiled-coil in the C-terminus, the eight TRPM members are clustered into four groups: TRPM1/M3, M2/M8, M4/M5 and M6/M7. TRPM subfamily members have been involved in several physiological functions. However, they are also linked to diverse pathophysiological human processes. Alterations in the expression and function of TRPM subfamily ion channels might generate several human diseases including cardiovascular and neurodegenerative alterations, organ dysfunction, cancer and many other channelopathies. These effects position them as remarkable putative targets for novel diagnostic strategies, drug design and therapeutic approaches. Here, we review the current knowledge about the main characteristics of all members of the TRPM family, focusing on their actions in human diseases.
Collapse
Affiliation(s)
- Ivanka Jimenez
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
| | - Yolanda Prado
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
| | - Felipe Marchant
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
| | - Carolina Otero
- Faculty of Medicine, School of Chemistry and Pharmacy, Universidad Andrés Bello, Santiago 8370186, Chile;
| | - Felipe Eltit
- Vancouver Prostate Centre, Vancouver, BC V6Z 1Y6, Canada;
- Department of Urological Sciences, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Claudio Cabello-Verrugio
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 7560484, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
| | - Oscar Cerda
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Felipe Simon
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
| |
Collapse
|
39
|
Prikhodko V, Chernyuk D, Sysoev Y, Zernov N, Okovityi S, Popugaeva E. Potential Drug Candidates to Treat TRPC6 Channel Deficiencies in the Pathophysiology of Alzheimer's Disease and Brain Ischemia. Cells 2020; 9:cells9112351. [PMID: 33114455 PMCID: PMC7692306 DOI: 10.3390/cells9112351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer’s disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer’s disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer’s disease and cerebral ischemia.
Collapse
Affiliation(s)
- Veronika Prikhodko
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Daria Chernyuk
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Yurii Sysoev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 St. Petersburg, Russia
| | - Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Sergey Okovityi
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Correspondence:
| |
Collapse
|
40
|
Zhang H, Zhao S, Yu J, Yang W, Liu Z, Zhang L. Medicinal chemistry perspective of TRPM2 channel inhibitors: where we are and where we might be heading? Drug Discov Today 2020; 25:2326-2334. [PMID: 33065292 DOI: 10.1016/j.drudis.2020.09.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/26/2020] [Accepted: 09/30/2020] [Indexed: 01/06/2023]
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a Ca2+- permeable nonselective cation channel that is involved in diverse biological functions as a cellular sensor for oxidative stress and temperature. It has been considered a promising therapeutic target for the treatment of ischemia/reperfusion (IR) injury, inflammation, cancer, and neurodegenerative diseases. Development of highly potent and selective TRPM2 inhibitors and validation of their use in relevant disease models will advance drug discovery. In this review, we describe the molecular structures and gating mechanism of the TRPM2 channel, and offer a comprehensive review of advances in the discovery of TRPM2 inhibitors. Furthermore, we analyze the properties of reported TRPM2 inhibitors with an emphasis on how specific inhibitors targeting this channel could be better developed.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Siqi Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jie Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wei Yang
- Department of Biophysics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
41
|
Gacki M, Kafarska K, Pietrzak A, Szczesio M, Korona-Głowniak I, Wolf WM. Transition Metal Complexes with Flufenamic Acid for Pharmaceutical Applications-A Novel Three-Centered Coordination Polymer of Mn(II) Flufenamate. MATERIALS (BASEL, SWITZERLAND) 2020; 13:ma13173705. [PMID: 32825746 PMCID: PMC7503579 DOI: 10.3390/ma13173705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 06/11/2023]
Abstract
Five complexes of Mn(II), Co(II), Ni(II), Cu(II) and Zn(II) with non-steroidal anti-inflammatory drug, flufenamic acid were synthesized: (1) [Mn3(fluf)6EtOH)(H2O)]·3EtOH; (2) [Co(fluf)2(EtOH)(H2O)]·H2O; (3) [Ni(fluf)2(EtOH)(H2O)]·H2O; (4) [Cu(fluf)2·H2O]; (5) [Zn(fluf)2·H2O]. All complexes were characterized by elemental analysis (EA), flame atomic absorption spectrometry (FAAS), Fourier-transform infrared spectroscopy (FTIR), and thermogravimetric analysis (TGA). The crystal structure of 1 was determined by the single crystal X-ray diffraction technique. It crystallizes in the triclinic space group P with three independent Mn(II) cations, six coordinated flufenamato ligands augmented with water and ethanol molecules in the inner coordination sphere. In this crystal, manganese atoms are multiplied by symmetry and form infinite, polymeric chains which extend along the [001] dimension. The Hirshfeld Surface analysis revealed changes in interaction assemblies around all metal centers. The antioxidant and antimicrobial activities were established for all complexes and free ligand for comparison. All compounds exhibit good or moderate bioactivity against Gram-positive bacteria and yeasts.
Collapse
Affiliation(s)
- Michał Gacki
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromskiego Street, 90–924 Lodz, Poland; (K.K.); (A.P.); (M.S.); (W.M.W.)
| | - Karolina Kafarska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromskiego Street, 90–924 Lodz, Poland; (K.K.); (A.P.); (M.S.); (W.M.W.)
| | - Anna Pietrzak
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromskiego Street, 90–924 Lodz, Poland; (K.K.); (A.P.); (M.S.); (W.M.W.)
| | - Małgorzata Szczesio
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromskiego Street, 90–924 Lodz, Poland; (K.K.); (A.P.); (M.S.); (W.M.W.)
| | - Izabela Korona-Głowniak
- Department of Pharmaceutical Microbiology, Medical University of Lublin, Chodzki 1, 20–093 Lublin, Poland;
| | - Wojciech M. Wolf
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, 116 Zeromskiego Street, 90–924 Lodz, Poland; (K.K.); (A.P.); (M.S.); (W.M.W.)
| |
Collapse
|
42
|
Mazzarda F, D'Elia A, Massari R, De Ninno A, Bertani FR, Businaro L, Ziraldo G, Zorzi V, Nardin C, Peres C, Chiani F, Tettey-Matey A, Raspa M, Scavizzi F, Soluri A, Salvatore AM, Yang J, Mammano F. Organ-on-chip model shows that ATP release through connexin hemichannels drives spontaneous Ca 2+ signaling in non-sensory cells of the greater epithelial ridge in the developing cochlea. LAB ON A CHIP 2020; 20:3011-3023. [PMID: 32700707 DOI: 10.1039/d0lc00427h] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Prior work supports the hypothesis that ATP release through connexin hemichannels drives spontaneous Ca2+ signaling in non-sensory cells of the greater epithelial ridge (GER) in the developing cochlea; however, direct proof is lacking. To address this issue, we plated cochlear organotypic cultures (COCs) and whole cell-based biosensors with nM ATP sensitivity (ATP-WCBs) at the bottom and top of an ad hoc designed transparent microfluidic chamber, respectively. By performing dual multiphoton Ca2+ imaging, we monitored the propagation of intercellular Ca2+ waves in the GER of COCs and ATP-dependent Ca2+ responses in overlying ATP-WCBs. Ca2+ signals in both COCs and ATP-WCBs were inhibited by supplementing the extracellular medium with ATP diphosphohydrolase (apyrase). Spontaneous Ca2+ signals were strongly depressed in the presence of Gjb6-/- COCs, in which connexin 30 (Cx30) is absent and connexin 26 (Cx26) is strongly downregulated. In contrast, spontaneous Ca2+ signals were not affected by replacement of Panx1-/- with Panx1+/+ COCs in the microfluidic chamber. Similar results were obtained by estimating ATP release from COCs using a classical luciferin-luciferase bioluminescence assay. Therefore, connexin hemichannels and not pannexin 1 channels mediate the release of ATP that is responsible for Ca2+ wave propagation in the developing mouse cochlea. The technological advances presented here have the potential to shed light on a plethora of unrelated open issues that involve paracrine signaling in physiology and pathology and cannot be addressed with standard methods.
Collapse
Affiliation(s)
- Flavia Mazzarda
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy. and Department of Science, Università degli Studi di Roma3, Rome, Italy
| | - Annunziata D'Elia
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy. and Department of Science, Università degli Studi di Roma3, Rome, Italy
| | - Roberto Massari
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy.
| | - Adele De Ninno
- CNR Institute for Photonics and Nanotechnology, Rome, Italy.
| | | | - Luca Businaro
- CNR Institute for Photonics and Nanotechnology, Rome, Italy.
| | - Gaia Ziraldo
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy. and Institute of Otorhinolaryngology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Veronica Zorzi
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy. and Institute of Otorhinolaryngology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara Nardin
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy.
| | - Chiara Peres
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy.
| | - Francesco Chiani
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy.
| | | | - Marcello Raspa
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy.
| | | | - Alessandro Soluri
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy.
| | | | - Jun Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. and Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Fabio Mammano
- CNR Institute of Biochemistry and Cell Biology, Monterotondo, Rome, Italy. and Department of Physics and Astronomy "G. Galilei", University of Padova, Padua, Italy.
| |
Collapse
|
43
|
Du EJ, Kang K. A Single Natural Variation Determines Cytosolic Ca 2+-Mediated Hyperthermosensitivity of TRPA1s from Rattlesnakes and Boas. Mol Cells 2020; 43:572-580. [PMID: 32484163 PMCID: PMC7332359 DOI: 10.14348/molcells.2020.0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/21/2020] [Accepted: 05/11/2020] [Indexed: 11/27/2022] Open
Abstract
Transient receptor potential ankyrin 1 from rattlesnakes (rsTRPA1) and boas (bTRPA1) was previously proposed to underlie thermo-sensitive infrared sensing based on transcript enrichment in infrared-sensing neurons and hyper-thermosensitivity expressed in Xenopus oocytes. It is unknown how these TRPA1s show thermosensitivities that overwhelm other thermoreceptors, and why rsTRPA1 is more thermosensitive than bTRPA1. Here, we show that snake TRPA1s differentially require Ca2+ for hyper-thermosensitivity and that predisposition to cytosolic Ca2+ potentiation correlates with superior thermosensitivity. Extracellularly applied Ca2+ upshifted the temperature coefficients (Q10s) of both TRPA1s, for which rsTRPA1, but not bTRPA1, requires cytosolic Ca2+. Intracellular Ca2+ chelation and substitutive mutations of the conserved cytosolic Ca2+-binding domain lowered rsTRPA1 thermosensitivity comparable to that of bTRPA1. Thapsigargin-evoked Ca2+ or calmodulin little affected rsTRPA1 activity or thermosensitivity, implying the importance of precise spatiotemporal action of Ca2+. Remarkably, a single rattlesnake-mimicking substitution in the conserved but presumably dormant cytosolic Ca2+-binding domain of bTRPA1 substantially enhanced thermosensitivity through cytosolic Ca2+ like rsTRPA1, indicating the capability of this single site in the determination of both cytosolic Ca2+ dependence and thermosensitivity. Collectively, these data suggest that Ca2+ is essential for the hyper-thermosensitivity of these TRPA1s, and cytosolic potentiation by permeating Ca2+ may contribute to the natural variation of infrared senses between rattlesnakes and boas.
Collapse
Affiliation(s)
- Eun Jo Du
- Department of Anatomy and Cell Biology and Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - KyeongJin Kang
- Department of Anatomy and Cell Biology and Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| |
Collapse
|
44
|
Bal R, Ozturk G, Etem EO, Eraslan E, Ozaydin S. Modulation of the excitability of stellate neurons in the ventral cochlear nucleus of mice by TRPM2 channels. Eur J Pharmacol 2020; 882:173163. [PMID: 32485244 DOI: 10.1016/j.ejphar.2020.173163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/14/2020] [Accepted: 04/29/2020] [Indexed: 10/24/2022]
Abstract
Oxidative stress-induced Ca2+ permeable transient receptor potential melastatin 2 (TRPM2) channels are expressed at high levels in the brain, appear to link neuronal excitability to cellular metabolism, and are involved in the pathogenesis of neurodegenerative disorders. We aimed to study the electrophysiological properties of TRPM2 channels in stellate cells of the mouse ventral cochlear nucleus (VCN) using molecular, immunohistochemical and electrophysiological approaches. In the present study, the real time PCR analysis revealed the presence of the TRPM2 mRNA in the mouse VCN tissue. Cell bodies of stellate cells were moderately labeled with TRPM2 antibodies using immunohistochemical staining. Stellate cells were sensitive to intracellular ADP-ribose (ADPR), a TRPM2 agonist. Upon the application of ADPR, the resting membrane potential of the stellate cells was significantly depolarized, shifting from -61.2 ± 0.9 mV to -57.0 ± 0.8 mV (P < 0.001; n = 21), and the firing rate significantly increased (P < 0.001, n = 6). When the pipette solution contained ADPR (300 μM) and the TRPM2 antagonists flufenamic acid (FFA) (100 μM), N-(p-amylcinnamoyl) anthranilic acid (ACA) (50 μM) and 8-bromo-cADP-Ribose (8-Br-cADPR) (50 μM), the membrane potential shifted in a hyperpolarizing direction. ADPR did not significantly change the resting membrane potential and action potential firing rate of stellate cells from TRPM2-/- mice. In conclusion, the results obtained using these molecular, immunohistochemical and electrophysiological approaches reveal the expression of functional TRPM2 channels in stellate neurons of the mouse VCN. TRPM2 might exert a significant modulatory effect on setting the level of resting excitability.
Collapse
Affiliation(s)
- Ramazan Bal
- Dept. of Physiology, Faculty of Medicine, Gaziantep University, 27310, Gaziantep, Turkey.
| | - Gurkan Ozturk
- Department of Physiology, Faculty of Medicine, Medipol University, Istanbul, Turkey
| | - Ebru Onalan Etem
- Dept. of Medical Biology, Faculty of Medicine, Firat University, 23119, Elazig, Turkey
| | - Ersen Eraslan
- Dept. of Physiology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Seda Ozaydin
- Dept. of Medical Biology, Faculty of Medicine, Firat University, 23119, Elazig, Turkey
| |
Collapse
|
45
|
Buntschu S, Tscherter A, Heidemann M, Streit J. Critical Components for Spontaneous Activity and Rhythm Generation in Spinal Cord Circuits in Culture. Front Cell Neurosci 2020; 14:81. [PMID: 32410961 PMCID: PMC7198714 DOI: 10.3389/fncel.2020.00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/19/2020] [Indexed: 11/23/2022] Open
Abstract
Neuronal excitability contributes to rhythm generation in central pattern generating networks (CPGs). In spinal cord CPGs, such intrinsic excitability partly relies on persistent sodium currents (INaP), whereas respiratory CPGs additionally depend on calcium-activated cation currents (ICAN). Here, we investigated the contributions of INaP and ICAN to spontaneous rhythm generation in neuronal networks of the spinal cord and whether they mainly involve Hb9 neurons. We used cultures of ventral and transverse slices from the E13-14 embryonic rodent lumbar spinal cord on multielectrode arrays (MEAs). All cultures showed spontaneous bursts of network activity. Blocking synaptic excitation with the AMPA receptor antagonist CNQX suppressed spontaneous network bursts and left asynchronous intrinsic activity at about 30% of the electrodes. Such intrinsic activity was completely blocked at all electrodes by both the INaP blocker riluzole as well as by the ICAN blocker flufenamic acid (FFA) and the more specific TRPM4 channel antagonist 9-phenanthrol. All three antagonists also suppressed spontaneous bursting completely and strongly reduced stimulus-evoked bursts. Also, FFA reduced repetitive spiking that was induced in single neurons by injection of depolarizing current pulses to few spikes. Other antagonists of unspecific cation currents or calcium currents had no suppressing effects on either intrinsic activity (gadolinium chloride) or spontaneous bursting (the TRPC channel antagonists clemizole and ML204 and the T channel antagonist TTA-P2). Combined patch-clamp and MEA recordings showed that Hb9 interneurons were activated by network bursts but could not initiate them. Together these findings suggest that both INaP through Na+-channels and ICAN through putative TRPM4 channels contribute to spontaneous intrinsic and repetitive spiking in spinal cord neurons and thereby to the generation of network bursts.
Collapse
Affiliation(s)
| | | | | | - Jürg Streit
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
46
|
Basu M, Gupta P, Dutta A, Jana K, Ukil A. Increased host ATP efflux and its conversion to extracellular adenosine is crucial for establishing Leishmania infection. J Cell Sci 2020; 133:jcs239939. [PMID: 32079656 DOI: 10.1242/jcs.239939] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Intracellular survival of Leishmania donovani demands rapid production of host ATP for its sustenance. However, a gradual decrease in intracellular ATP in spite of increased glycolysis suggests ATP efflux during infection. Accordingly, upon infection, we show here that ATP is exported and the major exporter was pannexin-1, leading to raised extracellular ATP levels. Extracellular ATP shows a gradual decrease after the initial increase, and analysis of cell surface ATP-degrading enzymes revealed induction of the ectonucleotidases CD39 and CD73. Ectonucleotidase-mediated ATP degradation leads to increased extracellular adenosine (eADO), and inhibition of CD39 and CD73 in infected cells decreased adenosine concentration and parasite survival, documenting the importance of adenosine in infection. Inhibiting adenosine uptake by cells did not affect parasite survival, suggesting that eADO exerts its effect through receptor-mediated signalling. We also show that Leishmania induces the expression of adenosine receptors A2AR and A2BR, both of which are important for anti-inflammatory responses. Treating infected BALB/c mice with CD39 and CD73 inhibitors resulted in decreased parasite burden and increased host-favourable cytokine production. Collectively, these observations indicate that infection-induced ATP is exported, and after conversion into adenosine, propagates infection via receptor-mediated signalling.
Collapse
Affiliation(s)
- Moumita Basu
- Department of Biochemistry, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Purnima Gupta
- Infections and Cancer Biology Group, International Agency for Research on Cancer, 69372, Lyon Cedex 08, France
| | - Ananya Dutta
- Division of Molecular Medicine, Bose Institute, P1/12 Calcutta Improvement Trust Scheme, VIIM, Kolkata, 700054, West Bengal, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, P1/12 Calcutta Improvement Trust Scheme, VIIM, Kolkata, 700054, West Bengal, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata 700019, West Bengal, India
| |
Collapse
|
47
|
Inhibition of Fast Nerve Conduction Produced by Analgesics and Analgesic Adjuvants-Possible Involvement in Pain Alleviation. Pharmaceuticals (Basel) 2020; 13:ph13040062. [PMID: 32260535 PMCID: PMC7243109 DOI: 10.3390/ph13040062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023] Open
Abstract
Nociceptive information is transmitted from the periphery to the cerebral cortex mainly by action potential (AP) conduction in nerve fibers and chemical transmission at synapses. Although this nociceptive transmission is largely inhibited at synapses by analgesics and their adjuvants, it is possible that the antinociceptive drugs inhibit nerve AP conduction, contributing to their antinociceptive effects. Many of the drugs are reported to inhibit the nerve conduction of AP and voltage-gated Na+ and K+ channels involved in its production. Compound action potential (CAP) is a useful measure to know whether drugs act on nerve AP conduction. Clinically-used analgesics and analgesic adjuvants (opioids, non-steroidal anti-inflammatory drugs, 2-adrenoceptor agonists, antiepileptics, antidepressants and local anesthetics) were found to inhibit fast-conducting CAPs recorded from the frog sciatic nerve by using the air-gap method. Similar actions were produced by antinociceptive plant-derived chemicals. Their inhibitory actions depended on the concentrations and chemical structures of the drugs. This review article will mention the inhibitory actions of the antinociceptive compounds on CAPs in frog and mammalian peripheral (particularly, sciatic) nerves and on voltage-gated Na+ and K+ channels involved in AP production. Nerve AP conduction inhibition produced by analgesics and analgesic adjuvants is suggested to contribute to at least a part of their antinociceptive effects.
Collapse
|
48
|
Dissolution Behavior of Flufenamic Acid in Heated Mixtures with Nanocellulose. Molecules 2020; 25:molecules25061277. [PMID: 32168901 PMCID: PMC7144018 DOI: 10.3390/molecules25061277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 11/16/2022] Open
Abstract
Flufenamic acid (FFA) is a problem drug that has up to eight different polymorphs and shows poor solubility. Variability in bioavailability has been reported in the past resulting in limited use of FFA in the oral solid dosage form. The goal of this article was to investigate the polymorphism and amorphization behavior of FFA in non-heated and heated mixtures with high surface area nanocellulose, i.e., Cladophora cellulose (CLAD). As a benchmark, low surface area microcrystalline cellulose (MCC) was used. The solid-state properties of mixtures were characterized with X-ray diffraction, Fourier-transform infrared spectroscopy, and differential scanning calorimetry. The dissolution behavior of mixtures was studied in three biorelevant media, i.e., fasted state simulated gastric fluid, fasted state simulated intestinal fluid, and fed state simulated intestinal fluid. Additional thermal analysis and dissolution tests were carried out following 4 months of storage at 75% RH and room temperature. Heated mixtures of FFA with CLAD resulted in complete amorphization of the drug, whereas that with MCC produced a mixture of up to four different polymorphs. The amorphous FFA mixture with CLAD exhibited rapid and invariable fasted/fed state dissolution in simulated intestinal fluids, whereas that of MCC mixtures was highly dependent on the biorelevant medium. The storage of the heated FFA-CLAD mixture did not result in recrystallization or changes in dissolution profile, whereas heated FFA-MCC mixture showed polymorphic changes. The straightforward dry powder formulation strategy presented here bears great promise for reformulating a number of problem drugs to enhance their dissolution properties and reduce the fasted/fed state variability.
Collapse
|
49
|
Maestrelli F, Rossi P, Paoli P, De Luca E, Mura P. The role of solid state properties on the dissolution performance of flufenamic acid. J Pharm Biomed Anal 2020; 180:113058. [PMID: 31881398 DOI: 10.1016/j.jpba.2019.113058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 10/25/2022]
Abstract
Flufenamic acid is a nonsteroidal anti-inflammatory drug characterized by a low solubility and a variable oral bioavailability. Flufenamic acid is present in the commercial products in two polymorphic enantiotropic forms (Form I and III). Bioinequivalence was observed for commercial solid dosage forms due to the different dissolution rate of batches. Aim of this work is the full characterization of the solid state properties of flufenamic acid in order to evidence reasons of its variable dissolution properties. Two different batches of pure drug obtained by different suppliers were fully characterized. In order to evaluate the effect of the technological processes used for tablet production, the powders were submitted to grinding, kneading, and compression. Thermal analysis and X-ray diffraction studies proved that the drug was provided by both suppliers as Form I, Form III is obtained by recrystallization from ethanol or ethanol/water of both batches and no changes were observed after the different mechanical treatments. No difference was observed between the two forms in terms of equilibrium solubility values. Dissolution rate studies evidenced a difference between the two batches due to their different particle size, which disappeared after sieving. Interestingly, a significant difference in terms of intrinsic dissolution rate and surface wettability of the two compacted powders was observed, even after sieving, probably related to a different behavior of the two powder samples under compaction. These results should be taken into account, during a tablet formulation, in order to obtain a reproducible dissolution performance of the drug, regardless of its original supplier.
Collapse
Affiliation(s)
- Francesca Maestrelli
- Department of Chemistry "U. Schiff", University of Florence, via U. Schiff, 6 Sesto Fiorentino, Florence, Italy.
| | - Patrizia Rossi
- Department of Industrial Engineering, University of Florence, via Santa Marta 3, Florence, Italy.
| | - Paola Paoli
- Department of Industrial Engineering, University of Florence, via Santa Marta 3, Florence, Italy.
| | - Enrico De Luca
- Department of Chemistry "U. Schiff", University of Florence, via U. Schiff, 6 Sesto Fiorentino, Florence, Italy.
| | - Paola Mura
- Department of Chemistry "U. Schiff", University of Florence, via U. Schiff, 6 Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
50
|
Involvement of TRPC4 and 5 Channels in Persistent Firing in Hippocampal CA1 Pyramidal Cells. Cells 2020; 9:cells9020365. [PMID: 32033274 PMCID: PMC7072216 DOI: 10.3390/cells9020365] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/29/2020] [Accepted: 02/01/2020] [Indexed: 12/17/2022] Open
Abstract
Persistent neural activity has been observed in vivo during working memory tasks, and supports short-term (up to tens of seconds) retention of information. While synaptic and intrinsic cellular mechanisms of persistent firing have been proposed, underlying cellular mechanisms are not yet fully understood. In vitro experiments have shown that individual neurons in the hippocampus and other working memory related areas support persistent firing through intrinsic cellular mechanisms that involve the transient receptor potential canonical (TRPC) channels. Recent behavioral studies demonstrating the involvement of TRPC channels on working memory make the hypothesis that TRPC driven persistent firing supports working memory a very attractive one. However, this view has been challenged by recent findings that persistent firing in vitro is unchanged in TRPC knock out (KO) mice. To assess the involvement of TRPC channels further, we tested novel and highly specific TRPC channel blockers in cholinergically induced persistent firing in mice CA1 pyramidal cells for the first time. The application of the TRPC4 blocker ML204, TRPC5 blocker clemizole hydrochloride, and TRPC4 and 5 blocker Pico145, all significantly inhibited persistent firing. In addition, intracellular application of TRPC4 and TRPC5 antibodies significantly reduced persistent firing. Taken together these results indicate that TRPC4 and 5 channels support persistent firing in CA1 pyramidal neurons. Finally, we discuss possible scenarios causing these controversial observations on the role of TRPC channels in persistent firing.
Collapse
|