413 results on '"TRPM Cation Channels physiology"'
Search Results
102. TRPM8 is a neuronal osmosensor that regulates eye blinking in mice.
- Author
-
Quallo T, Vastani N, Horridge E, Gentry C, Parra A, Moss S, Viana F, Belmonte C, Andersson DA, and Bevan S
- Subjects
- Action Potentials, Animals, CHO Cells, Cold Temperature, Cornea physiology, Cricetinae, Cricetulus, Female, Male, Mice, Mice, Knockout, Osmolar Concentration, Blinking, Sensory Receptor Cells physiology, TRPM Cation Channels physiology
- Abstract
Specific peripheral sensory neurons respond to increases in extracellular osmolality but the mechanism responsible for excitation is unknown. Here we show that small increases in osmolality excite isolated mouse dorsal root ganglion (DRG) and trigeminal ganglion (TG) neurons expressing the cold-sensitive TRPM8 channel (transient receptor potential channel, subfamily M, member 8). Hyperosmotic responses were abolished by TRPM8 antagonists, and were absent in DRG and TG neurons isolated from Trpm8(-/-) mice. Heterologously expressed TRPM8 was activated by increased osmolality around physiological levels and inhibited by reduced osmolality. Electrophysiological studies in a mouse corneal preparation demonstrated that osmolality regulated the electrical activity of TRPM8-expressing corneal afferent neurons. Finally, the frequency of eye blinks was reduced in Trpm8(-/-) compared with wild-type mice and topical administration of a TRPM8 antagonist reduced blinking in wild-type mice. Our findings identify TRPM8 as a peripheral osmosensor responsible for the regulation of normal eye-blinking in mice.
- Published
- 2015
- Full Text
- View/download PDF
103. [Role of transient receptor potential melastatin 8 channels in migraine mechanism in rats].
- Author
-
Qin DM, Zou Z, Zhou CR, and Mu FG
- Subjects
- Animals, Cyclic AMP-Dependent Protein Kinases analysis, Male, Rats, Rats, Sprague-Dawley, Receptors, N-Methyl-D-Aspartate analysis, Receptors, N-Methyl-D-Aspartate physiology, Migraine Disorders etiology, TRPM Cation Channels physiology
- Abstract
Objective: To investigate the role of transient receptor potential melastatin 8 (TRPM8) channels in migraine mechanism in rats by measuring the changes in expression of TRPM8 in the trigeminal nerve of rats with migraine., Methods: Twenty male Sprague-Dawley rats were randomly and equally divided into a blank control group and a model group. Nitroglycerin (10 mg/kg) was injected subcutaneously in the back of the neck once a week for 5 weeks, to prepared a rat model of migraine without aura. Normal saline was injected subcutaneously instead of nitroglycerin in the control group. At 4 hours after the final injection, behavior scoring of all rats was performed, and then the trigeminal nerve ganglions of rats in both groups were collected for measurement of expression of N-methyl-D-aspartate receptor (NMDAR), protein kinase A (PKA), and TRPM8 using immunohistochemical staining, immunofluorescence, and Western blot, respectively., Results: The behavior score in each week during the rat model preparing was significantly higher in the model group than in the control group (P<0.05). The expression of NMDAR, PKA, and TRPM8 in the model group was significantly higher than in the control group (P<0.01). Both the behavior score and the expression of NMDAR were positively correlated with the expression of TRPM8 (r=0.822 and 0.794 respectively; P<0.01)., Conclusions: TRPM8 may be involved in migraine mechanism probably by activation of the NMDAR pathway.
- Published
- 2015
104. TRPM7 maintains progenitor-like features of neuroblastoma cells: implications for metastasis formation.
- Author
-
Middelbeek J, Visser D, Henneman L, Kamermans A, Kuipers AJ, Hoogerbrugge PM, Jalink K, and van Leeuwen FN
- Subjects
- Animals, Bone Marrow Neoplasms secondary, Cell Division, Cell Line, Tumor, Cell Movement, Disease Progression, Gene Expression Regulation, Developmental, Gene Expression Regulation, Neoplastic, Heterografts, Humans, Liver Neoplasms secondary, Mice, Neural Crest cytology, Neuroblastoma metabolism, RNA Interference, RNA, Small Interfering genetics, Signal Transduction genetics, Snail Family Transcription Factors, Transcription Factors physiology, Transcription, Genetic, Tumor Microenvironment, Neoplasm Metastasis genetics, Neoplasm Proteins physiology, Neoplastic Stem Cells cytology, Neuroblastoma pathology, Protein Serine-Threonine Kinases physiology, TRPM Cation Channels physiology
- Abstract
Neuroblastoma is an embryonal tumor derived from poorly differentiated neural crest cells. Current research is aimed at identifying the molecular mechanisms that maintain the progenitor state of neuroblastoma cells and to develop novel therapeutic strategies that induce neuroblastoma cell differentiation. Mechanisms controlling neural crest development are typically dysregulated during neuroblastoma progression, and provide an appealing starting point for drug target discovery. Transcriptional programs involved in neural crest development act as a context dependent gene regulatory network. In addition to BMP, Wnt and Notch signaling, activation of developmental gene expression programs depends on the physical characteristics of the tissue microenvironment. TRPM7, a mechanically regulated TRP channel with kinase activity, was previously found essential for embryogenesis and the maintenance of undifferentiated neural crest progenitors. Hence, we hypothesized that TRPM7 may preserve progenitor-like, metastatic features of neuroblastoma cells. Using multiple neuroblastoma cell models, we demonstrate that TRPM7 expression closely associates with the migratory and metastatic properties of neuroblastoma cells in vitro and in vivo. Moreover, microarray-based expression profiling on control and TRPM7 shRNA transduced neuroblastoma cells indicates that TRPM7 controls a developmental transcriptional program involving the transcription factor SNAI2. Overall, our data indicate that TRPM7 contributes to neuroblastoma progression by maintaining progenitor-like features.
- Published
- 2015
- Full Text
- View/download PDF
105. TRPM2-mediated intracellular Zn2+ release triggers pancreatic β-cell death.
- Author
-
Manna PT, Munsey TS, Abuarab N, Li F, Asipu A, Howell G, Sedo A, Yang W, Naylor J, Beech DJ, Jiang LH, and Sivaprasadarao A
- Subjects
- Animals, Cell Death physiology, HEK293 Cells, Humans, Insulin-Secreting Cells pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Reactive Oxygen Species metabolism, Insulin-Secreting Cells metabolism, Intracellular Fluid metabolism, TRPM Cation Channels physiology, Zinc metabolism
- Abstract
Reactive oxygen species (ROS) can cause pancreatic β-cell death by activating transient receptor potential (melastatin) 2 (TRPM2) channels. Cell death has been attributed to the ability of these channels to raise cytosolic Ca2+. Recent studies however revealed that TRPM2 channels can also conduct Zn2+, but the physiological relevance of this property is enigmatic. Given that Zn2+ is cytotoxic, we asked whether TRPM2 channels can permeate sufficient Zn2+ to affect cell viability. To address this, we used the insulin secreting (INS1) β-cell line, human embryonic kidney (HEK)-293 cells transfected with TRPM2 and pancreatic islets. H2O2 activation of TRPM2 channels increases the cytosolic levels of both Ca2+ and Zn2+ and causes apoptotic cell death. Interestingly, chelation of Zn2+ alone was sufficient to prevent β-cell death. The source of the cytotoxic Zn2+ is intracellular, found largely sequestered in lysosomes. Lysosomes express TRPM2 channels, providing a potential route for Zn2+ release. Zn2+ release is potentiated by extracellular Ca2+ entry, indicating that Ca2+-induced Zn2+ release leads to apoptosis. Knockout of TRPM2 channels protects mice from β-cell death and hyperglycaemia induced by multiple low-dose streptozotocin (STZ; MLDS) administration. These results argue that TRPM2-mediated, Ca2+-potentiated Zn2+ release underlies ROS-induced β-cell death and Zn2+, rather than Ca2+, plays a primary role in apoptosis.
- Published
- 2015
- Full Text
- View/download PDF
106. Involvement of TRPM2 in a wide range of inflammatory and neuropathic pain mouse models.
- Author
-
So K, Haraguchi K, Asakura K, Isami K, Sakimoto S, Shirakawa H, Mori Y, Nakagawa T, and Kaneko S
- Subjects
- Animals, Disease Models, Animal, Female, Male, Mice, Inbred C57BL, Mice, Knockout, TRPM Cation Channels deficiency, Inflammation genetics, Neuralgia genetics, TRPM Cation Channels physiology
- Abstract
Recent evidence suggests a role of transient receptor potential melastatin 2 (TRPM2) in immune and inflammatory responses. We previously reported that TRPM2 deficiency attenuated inflammatory and neuropathic pain in some pain mouse models, including formalin- or carrageenan-induced inflammatory pain, and peripheral nerve injury-induced neuropathic pain models, while it had no effect on the basal mechanical and thermal nociceptive sensitivities. In this study, we further explored the involvement of TRPM2 in various pain models using TRPM2-knockout mice. There were no differences in the chemonociceptive behaviors evoked by intraplantar injection of capsaicin or hydrogen peroxide between wildtype and TRPM2-knockout mice, while acetic acid-induced writhing behavior was significantly attenuated in TRPM2-knockout mice. In the postoperative incisional pain model, no difference in mechanical allodynia was observed between the two genotypes. By contrast, mechanical allodynia in the monosodium iodoacetate-induced osteoarthritis pain model and the experimental autoimmune encephalomyelitis model were significantly attenuated in TRPM2-knockout mice. Furthermore, mechanical allodynia in paclitaxel-induced peripheral neuropathy and streptozotocin-induced painful diabetic neuropathy models were significantly attenuated in TRPM2-knockout mice. Taken together, these results suggest that TRPM2 plays roles in a wide range of pathological pain models based on peripheral and central neuroinflammation, rather than physiological nociceptive pain., (Copyright © 2015 The Authors. Production and hosting by Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
107. Hypotonic stress induces RANKL via transient receptor potential melastatin 3 (TRPM3) and vaniloid 4 (TRPV4) in human PDL cells.
- Author
-
Son GY, Yang YM, Park WS, Chang I, and Shin DM
- Subjects
- Biomechanical Phenomena, Bone Remodeling physiology, Boron Compounds pharmacology, Calcium Channel Blockers pharmacology, Calcium Signaling drug effects, Calcium Signaling physiology, Cell Culture Techniques, Cells, Cultured, Gene Silencing, Humans, Hypotonic Solutions, Morpholines pharmacology, Osteoprotegerin biosynthesis, Periodontal Ligament cytology, Phorbols pharmacology, Pregnenolone pharmacology, Pyrroles pharmacology, RANK Ligand antagonists & inhibitors, RNA, Messenger metabolism, RNA, Small Interfering administration & dosage, Ruthenium Red pharmacology, Signal Transduction physiology, Stress, Mechanical, TRPM Cation Channels agonists, TRPM Cation Channels antagonists & inhibitors, TRPV Cation Channels agonists, TRPV Cation Channels antagonists & inhibitors, Periodontal Ligament metabolism, RANK Ligand biosynthesis, TRPM Cation Channels physiology, TRPV Cation Channels physiology
- Abstract
Bone remodeling occurs in response to various types of mechanical stress. The periodontal ligament (PDL) plays an important role in mechanical stress-mediated alveolar bone remodeling. However, the underlying mechanism at the cellular level has not been extensively studied. In this study, we investigated the effect of shear stress on the expression of bone remodeling factors, including receptor activator of nuclear factor-kappa B (NF-κB) ligand (RANKL) and osteoprotegerin (OPG), as well as its upstream signaling pathway in primary human PDL cells. We applied hypotonic stress to reproduce shear stress to PDL cells. Hypotonic stress induced the messenger RNA (mRNA) and protein expression of RANKL but not OPG. It also increased intracellular Ca(2+) concentration ([Ca(2+)]i). Extracellular Ca(2+) depletion and nonspecific plasma membrane Ca(2+) channel blockers completely inhibited the increase in both [Ca(2+)]i and RANKL mRNA expression. We identified the expression and activation of transient receptor potential melastatin 3 (TRPM3) and vaniloid 4 (TRPV4) channels in PDL cells. Pregnenolone sulfate (PS) and 4α-phorbol 12, 13-didecanoate (4α-PDD), which are agonists of TRPM3 and TRPV4, augmented Ca(2+) influx and RANKL mRNA expression. Both pharmacological (2-aminoethoxydiphenyl borate [2-APB], ruthenium red [RR], ononetin [Ono], and HC 067047 [HC]) and genetic (small interfering RNA [siRNA]) inhibitors of TRPM3 and TRPV4 reduced the hypotonic stress-mediated increase in [Ca(2+)]i and RANKL mRNA expression. Our study shows that hypotonic stress induced RANKL mRNA expression via TRPM3- and TRPV4-mediated extracellular Ca(2+) influx and RANKL expression. This signaling pathway in PDL cells may play a critical role in mechanical stress-mediated alveolar bone remodeling., (© International & American Associations for Dental Research 2015.)
- Published
- 2015
- Full Text
- View/download PDF
108. Reduction in traumatic brain injury-induced oxidative stress, apoptosis, and calcium entry in rat hippocampus by melatonin: Possible involvement of TRPM2 channels.
- Author
-
Yürüker V, Nazıroğlu M, and Şenol N
- Subjects
- Animals, Benzene Derivatives pharmacology, Biological Transport drug effects, Boron Compounds pharmacology, Brain Injuries metabolism, Capsaicin pharmacology, Caspases physiology, Drug Evaluation, Preclinical, Hippocampus metabolism, Male, Melatonin pharmacology, Membrane Potential, Mitochondrial drug effects, Neuroprotective Agents pharmacology, Random Allocation, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Apoptosis drug effects, Brain Injuries drug therapy, Calcium metabolism, Hippocampus drug effects, Melatonin therapeutic use, Nerve Tissue Proteins physiology, Neuroprotective Agents therapeutic use, Oxidative Stress drug effects, TRPM Cation Channels physiology
- Abstract
Melatonin, which is a very effective reactive oxygen species (ROS) scavenger, acts through a direct reaction with free radicals. Ca(2+) entry induced by traumatic brain injury (TBI) has deleterious effects on human hippocampal function. TRPM2 is a Ca(2+) permeable non-selective channel in hippocampal neurons, and its activation of during oxidative stress has been linked to cell death. Despite the importance of oxidative stress in TBI, its role in apoptosis and Ca(2+) entry in TBI is poorly understood. Therefore, we tested the effects of melatonin on apoptosis, oxidative stress, and Ca(2+) entry through the TRPM2 channel in the hippocampal neurons of TBI-induced rats. Thirty-two rats were divided into the following four groups: control, melatonin, TBI, and TBI + melatonin groups. Melatonin (5 mg/kg body weight) was intraperitoneally given to animals in the melatonin group and the TBI + melatonin group after 1 h of brain trauma. Hippocampal neurons were freshly isolated from the four groups, incubated with a nonspecific TRPM2 blocker (2-aminoethyl diphenylborinate, 2-APB), and then stimulated with cumene hydroperoxide. Apoptosis, caspase-3, caspase-9, intracellular ROS production, mitochondrial membrane depolarization and intracellular free Ca(2+) ([Ca(2+)]i) values were high in the TBI group, and low in the TBI + melatonin group. The [Ca(2+)]i concentration was decreased in the four groups by 2-APB. In our TBI experimental model, TRPM2 channels were involved in Ca(2+) entry-induced neuronal death, and the negative modulation of the activity of this channel by melatonin pretreatment may account for the neuroprotective activity of TRPM2 channels against oxidative stress, apoptosis, and Ca(2+) entry.
- Published
- 2015
- Full Text
- View/download PDF
109. The Ca(2+)-activated cation channel TRPM4 is a negative regulator of angiotensin II-induced cardiac hypertrophy.
- Author
-
Kecskés M, Jacobs G, Kerselaers S, Syam N, Menigoz A, Vangheluwe P, Freichel M, Flockerzi V, Voets T, and Vennekens R
- Subjects
- Animals, Animals, Newborn, Calcium metabolism, Male, Mice, Mice, Inbred C57BL, Myocytes, Cardiac metabolism, Phosphoric Monoester Hydrolases metabolism, Angiotensin II pharmacology, Cardiomegaly chemically induced, TRPM Cation Channels physiology
- Abstract
Cardiac muscle adapts to hemodynamic stress by altering myocyte size and function, resulting in cardiac hypertrophy. Alteration in myocyte calcium homeostasis is known to be an initial signal in cardiac hypertrophy signaling. Transient receptor potential melastatin 4 protein (TRPM4) is a calcium-activated non-selective cation channel, which plays a role in regulating calcium influx and calcium-dependent cell functions in many cell types including cardiomyocytes. Selective deletion of TRPM4 from the heart muscle in mice resulted in an increased hypertrophic growth after chronic angiotensin (AngII) treatment, compared to WT mice. The enhanced hypertrophic response was also traceable by the increased expression of hypertrophy-related genes like Rcan1, ANP, and α-Actin. Intracellular calcium measurements on isolated ventricular myocytes showed significantly increased store-operated calcium entry upon AngII treatment in myocytes lacking the TRPM4 channel. Elevated intracellular calcium is a key factor in the development of pathological cardiac hypertrophy, leading to the activation of intracellular signaling pathways. In agreement with this, we observed significantly higher Rcan1 mRNA level, calcineurin enzyme activity and protein level in lysates from TRPM4-deficient mice heart compared to WT after AngII treatment. Collectively, these observations are consistent with a model in which TRPM4 is a regulator of calcium homeostasis in cardiomyocytes after AngII stimulation. TRPM4 contributes to the regulation of driving force for store-operated calcium entry and thereby the activation of the calcineurin-NFAT pathway and the development of pathological hypertrophy.
- Published
- 2015
- Full Text
- View/download PDF
110. A splice variant of the human ion channel TRPM2 modulates neuroblastoma tumor growth through hypoxia-inducible factor (HIF)-1/2α.
- Author
-
Chen SJ, Hoffman NE, Shanmughapriya S, Bao L, Keefer K, Conrad K, Merali S, Takahashi Y, Abraham T, Hirschler-Laszkiewicz I, Wang J, Zhang XQ, Song J, Barrero C, Shi Y, Kawasawa YI, Bayerl M, Sun T, Barbour M, Wang HG, Madesh M, Cheung JY, and Miller BA
- Subjects
- Adrenal Glands metabolism, Animals, Antibiotics, Antineoplastic pharmacology, Autophagy, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Line, Tumor, Cell Proliferation, Cell Survival drug effects, Down-Regulation, Doxorubicin pharmacology, Female, Gene Expression Regulation, Neoplastic, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Membrane Potential, Mitochondrial, Membrane Potentials, Mice, Nude, Neoplasm Transplantation, Neuroblastoma pathology, Protein Isoforms physiology, Protein Transport, Tumor Burden, Basic Helix-Loop-Helix Transcription Factors metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Neuroblastoma metabolism, TRPM Cation Channels physiology
- Abstract
The calcium-permeable ion channel TRPM2 is highly expressed in a number of cancers. In neuroblastoma, full-length TRPM2 (TRPM2-L) protected cells from moderate oxidative stress through increased levels of forkhead box transcription factor 3a (FOXO3a) and superoxide dismutase 2. Cells expressing the dominant negative short isoform (TRPM2-S) had reduced FOXO3a and superoxide dismutase 2 levels, reduced calcium influx in response to oxidative stress, and enhanced reactive oxygen species, leading to decreased cell viability. Here, in xenografts generated with SH-SY5Y neuroblastoma cells stably expressing TRPM2 isoforms, growth of tumors expressing TRPM2-S was significantly reduced compared with tumors expressing TRPM2-L. Expression of hypoxia-inducible factor (HIF)-1/2α was significantly reduced in TRPM2-S-expressing tumor cells as was expression of target proteins regulated by HIF-1/2α including those involved in glycolysis (lactate dehydrogenase A and enolase 2), oxidant stress (FOXO3a), angiogenesis (VEGF), mitophagy and mitochondrial function (BNIP3 and NDUFA4L2), and mitochondrial electron transport chain activity (cytochrome oxidase 4.1/4.2 in complex IV). The reduction in HIF-1/2α was mediated through both significantly reduced HIF-1/2α mRNA levels and increased levels of von Hippel-Lindau E3 ligase in TRPM2-S-expressing cells. Inhibition of TRPM2-L by pretreatment with clotrimazole or expression of TRPM2-S significantly increased sensitivity of cells to doxorubicin. Reduced survival of TRPM2-S-expressing cells after doxorubicin treatment was rescued by gain of HIF-1 or -2α function. These data suggest that TRPM2 activity is important for tumor growth and for cell viability and survival following doxorubicin treatment and that interference with TRPM2-L function may be a novel approach to reduce tumor growth through modulation of HIF-1/2α, mitochondrial function, and mitophagy., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
111. Negatively charged amino acids near and in transient receptor potential (TRP) domain of TRPM4 channel are one determinant of its Ca2+ sensitivity.
- Author
-
Yamaguchi S, Tanimoto A, Otsuguro K, Hibino H, and Ito S
- Subjects
- Animals, Aspartic Acid genetics, Binding Sites genetics, Calcium pharmacology, Cations, Divalent metabolism, Cations, Divalent pharmacology, Cobalt metabolism, Cobalt pharmacology, Glutamic Acid genetics, HEK293 Cells, Humans, Male, Membrane Potentials drug effects, Mutation, Patch-Clamp Techniques, Phosphatidylinositol 4,5-Diphosphate metabolism, Phosphatidylinositol 4,5-Diphosphate pharmacology, Rats, Inbred BN, TRPM Cation Channels genetics, TRPM Cation Channels physiology, Transfection, Aspartic Acid metabolism, Calcium metabolism, Glutamic Acid metabolism, TRPM Cation Channels metabolism
- Abstract
Transient receptor potential (TRP) channel melastatin subfamily member 4 (TRPM4) is a broadly expressed nonselective monovalent cation channel. TRPM4 is activated by membrane depolarization and intracellular Ca(2+), which is essential for the activation. The Ca(2+) sensitivity is known to be regulated by calmodulin and membrane phosphoinositides, such as phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). Although these regulators must play important roles in controlling TRPM4 activity, mutation analyses of the calmodulin-binding sites have suggested that Ca(2+) binds to TRPM4 directly. However, the intrinsic binding sites in TRPM4 remain to be elucidated. Here, by using patch clamp and molecular biological techniques, we show that there are at least two functionally different divalent cation-binding sites, and the negatively charged amino acids near and in the TRP domain in the C-terminal tail of TRPM4 (Asp-1049 and Glu-1062 of rat TRPM4) are required for maintaining the normal Ca(2+) sensitivity of one of the binding sites. Applications of Co(2+), Mn(2+), or Ni(2+) to the cytosolic side potentiated TRPM4 currents, increased the Ca(2+) sensitivity, but were unable to evoke TRPM4 currents without Ca(2+). Mutations of the acidic amino acids near and in the TRP domain, which are conserved in TRPM2, TRPM5, and TRPM8, deteriorated the Ca(2+) sensitivity in the presence of Co(2+) or PI(4,5)P2 but hardly affected the sensitivity to Co(2+) and PI(4,5)P2. These results suggest a novel role of the TRP domain in TRPM4 as a site responsible for maintaining the normal Ca(2+) sensitivity. These findings provide more insights into the molecular mechanisms of the regulation of TRPM4 by Ca(2+)., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
112. Temperature and voltage coupling to channel opening in transient receptor potential melastatin 8 (TRPM8).
- Author
-
Raddatz N, Castillo JP, Gonzalez C, Alvarez O, and Latorre R
- Subjects
- Algorithms, Animals, Cold Temperature, Electric Stimulation, Ion Channel Gating drug effects, Ion Channel Gating genetics, Kinetics, Membrane Potentials drug effects, Membrane Potentials physiology, Menthol pharmacology, Models, Biological, Oocytes metabolism, Oocytes physiology, Phosphatidylinositol 4,5-Diphosphate pharmacology, Rats, TRPM Cation Channels genetics, Xenopus laevis, Ion Channel Gating physiology, TRPM Cation Channels physiology, Temperature
- Abstract
Expressed in somatosensory neurons of the dorsal root and trigeminal ganglion, the transient receptor potential melastatin 8 (TRPM8) channel is a Ca(2+)-permeable cation channel activated by cold, voltage, phosphatidylinositol 4,5-bisphosphate, and menthol. Although TRPM8 channel gating has been characterized at the single channel and macroscopic current levels, there is currently no consensus regarding the extent to which temperature and voltage sensors couple to the conduction gate. In this study, we extended the range of voltages where TRPM8-induced ionic currents were measured and made careful measurements of the maximum open probability the channel can attain at different temperatures by means of fluctuation analysis. The first direct measurements of TRPM8 channel temperature-driven conformational rearrangements provided here suggest that temperature alone is able to open the channel and that the opening reaction is voltage-independent. Voltage is a partial activator of TRPM8 channels, because absolute open probability values measured with fully activated voltage sensors are less than 1, and they decrease as temperature rises. By unveiling the fast temperature-dependent deactivation process, we show that TRPM8 channel deactivation is well described by a double exponential time course. The fast and slow deactivation processes are temperature-dependent with enthalpy changes of 27.2 and 30.8 kcal mol(-1). The overall Q10 for the closing reaction is about 33. A three-tiered allosteric model containing four voltage sensors and four temperature sensors can account for the complex deactivation kinetics and coupling between voltage and temperature sensor activation and channel opening., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
113. Putative chanzyme activity of TRPM2 cation channel is unrelated to pore gating.
- Author
-
Tóth B, Iordanov I, and Csanády L
- Subjects
- Animals, Hydrolysis, Ligands, Xenopus laevis, Ion Channel Gating, TRPM Cation Channels physiology
- Abstract
Transient receptor potential melastatin 2 (TRPM2) is a Ca(2+)-permeable cation channel expressed in immune cells of phagocytic lineage, pancreatic β cells, and brain neurons and is activated under oxidative stress. TRPM2 activity is required for immune cell activation and insulin secretion and is responsible for postischemic neuronal cell death. TRPM2 is opened by binding of ADP ribose (ADPR) to its C-terminal cytosolic nudix-type motif 9 (NUDT9)-homology (NUDT9-H) domain, which, when expressed in isolation, cleaves ADPR into AMP and ribose-5-phosphate. A suggested coupling of this enzymatic activity to channel gating implied a potentially irreversible gating cycle, which is a unique feature of a small group of channel enzymes known to date. The significance of such a coupling lies in the conceptually distinct pharmacologic strategies for modulating the open probability of channels obeying equilibrium versus nonequilibrium gating mechanisms. Here we examine the potential coupling of TRPM2 enzymatic activity to pore gating. Mutation of several residues proposed to enhance or eliminate NUDT9-H catalytic activity all failed to affect channel gating kinetics. An ADPR analog, α-β-methylene-ADPR (AMPCPR), was shown to be entirely resistant to hydrolysis by NUDT9, but nevertheless supported TRPM2 channel gating, albeit with reduced apparent affinity. The rate of channel deactivation was not slowed but, rather, accelerated in AMPCPR. These findings, as well as detailed analyses of steady-state gating kinetics of single channels recorded in the presence of a range of concentrations of ADPR or AMPCPR, identify TRPM2 as a simple ligand-gated channel that obeys an equilibrium gating mechanism uncoupled from its enzymatic activity.
- Published
- 2014
- Full Text
- View/download PDF
114. Thermosensory signaling by TRPM is processed by brain serotonergic neurons to produce planarian thermotaxis.
- Author
-
Inoue T, Yamashita T, and Agata K
- Subjects
- Animals, Nerve Regeneration physiology, Synaptotagmins genetics, TRPM Cation Channels antagonists & inhibitors, Behavior, Animal physiology, Planarians physiology, Serotonergic Neurons physiology, Signal Transduction physiology, TRPM Cation Channels physiology, Thermosensing physiology
- Abstract
For most organisms, sensitive recognition of even slight changes in environmental temperature is essential for adjusting their behavioral strategies to ensure homeostasis and survival. However, much remains to be understood about the molecular and cellular processes that regulate thermosensation and the corresponding behavioral responses. Planarians display clear thermotaxis, although they have a relatively simple brain. Here, we devised a quantitative thermotaxis assay and unraveled a neural pathway involved in planarian thermotaxis by combinatory behavioral assays and RNAi analysis. We found that thermosensory neurons that expressed a planarian Dugesia japonica homolog of the Transient Receptor Potential Melastatin family a (DjTRPMa) gene were required for the thermotaxis. Interestingly, although these thermosensory neurons are distributed throughout their body, planarians with a dysfunctional brain due to regeneration-dependent conditional gene knockdown (Readyknock) of the synaptotagmin gene completely lost their thermotactic behavior. These results suggest that brain function is required as a central processor for the thermosensory response. Therefore, we investigated the type(s) of brain neurons involved in processing the thermal signals by gene knockdown of limiting enzymes for neurotransmitter biosynthesis in the brain. We found that serotonergic neurons with dendrites that were elongated toward DjTRPMa-expressing thermosensory neurons might be required for the processing of signals from thermosensory neurons that results in thermotaxis. These results suggest that serotonergic neurons in the brain may interact with thermosensory neurons activated by TRPM ion channels to produce thermotaxis in planarians., (Copyright © 2014 the authors 0270-6474/14/3415701-14$15.00/0.)
- Published
- 2014
- Full Text
- View/download PDF
115. Transient receptor potential melastatin subfamily member 2 cation channel regulates detrimental immune cell invasion in ischemic stroke.
- Author
-
Gelderblom M, Melzer N, Schattling B, Göb E, Hicking G, Arunachalam P, Bittner S, Ufer F, Herrmann AM, Bernreuther C, Glatzel M, Gerloff C, Kleinschnitz C, Meuth SG, Friese MA, and Magnus T
- Subjects
- Animals, Brain Ischemia pathology, Cells, Cultured, Hippocampus immunology, Hippocampus pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Organ Culture Techniques, Random Allocation, Stroke pathology, Brain Ischemia immunology, Cell Movement immunology, Immunity, Cellular immunology, Stroke immunology, TRPM Cation Channels physiology
- Abstract
Background and Purpose: Brain injury during stroke results in oxidative stress and the release of factors that include extracellular Ca(2+), hydrogen peroxide, adenosine diphosphate ribose, and nicotinic acid adenine dinucleotide phosphate. These alterations of the extracellular milieu change the activity of transient receptor potential melastatin subfamily member 2 (TRPM2), a nonselective cation channel expressed in the central nervous system and the immune system. Our goal was to evaluate the contribution of TRPM2 to the tissue damage after stroke., Methods: In accordance with current quality guidelines, we independently characterized Trpm2 in a murine ischemic stroke model in 2 different laboratories., Results: Gene deficiency of Trpm2 resulted in significantly improved neurological outcome and decreased infarct size. Besides an already known moderate neuroprotective effect of Trpm2 deficiency in vitro, ischemic brain invasion by neutrophils and macrophages was particularly reduced in Trpm2-deficient mice. Bone marrow chimeric mice revealed that Trpm2 deficiency in the peripheral immune system is responsible for the protective phenotype. Furthermore, experiments with mixed bone marrow chimeras demonstrated that Trpm2 is essential for the migration of neutrophils and, to a lesser extent, also of macrophages into ischemic hemispheres. Notably, the pharmacological TRPM2 inhibitor, N-(p-amylcinnamoyl)anthranilic acid, was equally protective in the stroke model., Conclusions: Although a neuroprotective effect of TRPM2 in vitro is well known, we can show for the first time that the detrimental role of TRPM2 in stroke primarily depends on its role in activating peripheral immune cells. Targeting TRPM2 systemically represents a promising therapeutic approach for ischemic stroke., (© 2014 American Heart Association, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
116. TGF-β1-elevated TRPM7 channel regulates collagen expression in hepatic stellate cells via TGF-β1/Smad pathway.
- Author
-
Fang L, Huang C, Meng X, Wu B, Ma T, Liu X, Zhu Q, Zhan S, and Li J
- Subjects
- Actins analysis, Animals, Collagen Type I analysis, Collagen Type I, alpha 1 Chain, Male, Rats, Rats, Sprague-Dawley, TRPM Cation Channels analysis, TRPM Cation Channels antagonists & inhibitors, Collagen metabolism, Hepatic Stellate Cells metabolism, Signal Transduction physiology, Smad Proteins physiology, TRPM Cation Channels physiology, Transforming Growth Factor beta1 physiology
- Abstract
Transdifferentiation of hepatic stellate cells (HSCs) into myofibroblasts plays a critical role in the development of liver fibrosis, since myofibroblasts are the key cells responsible for excessive deposition of ECM proteins. Transient receptor potential melastatin 7 (TRPM7), a non-selective cation channel with protein serine/threonine kinase activity, has been demonstrated to function in the proliferation of activated HSCs. Here, we investigated the functional role of TRPM7 in collagen deposition in activated HSC-T6 cells (a rat hepatic stellate cell line). TRPM7 mRNA and protein were measured by Real-time PCR and Western blot in TGF-β1-activated HSC-T6 cells in vitro. Results demonstrated that TRPM7 protein was dramatically increased in fibrotic human livers. Stimulation of HSC-T6 cells with TGF-β1 increased TRPM7 mRNA and protein level in a time-dependent manner. Nevertheless, TGF-β1-elicited upregulation of TRPM7 in HSC-T6 cells was abrogated by SB431542 (TGF-β1 receptor blocker) or SIS3 (inhibitor of Smad3 phosphorylation). Additionally, blockade of TRPM7 channels with non-specific TRPM7 blocker 2-APB or synthetic siRNA targeting TRPM7 attenuated TGF-β1-induced expression of myofibroblast markers, as measured by the induction of α-SMA and Col1α1. Silencing TRPM7 also increased the ratio of MMPs/TIMPs by increasing MMP-13 expression and decreasing TIMP-1 and TIMP-2 levels. Strikingly, phosphorylation of p-Smad2 and p-Smad3, associated with collagen production, was decreased in TRPM7 deficient HSC-T6 cells. These observations suggested that TGF-β1 elevates TRPM7 expression in HSCs via Smad3-dependant mechanisms, which in turn contributes Smad protein phosphorylation, and subsequently increases fibrous collagen expression. Therefore, TRPM7 may constitute a useful target for the treatment of liver fibrosis., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
117. The role of TRPM8 in the Guinea-pig bladder-cooling reflex investigated using a novel TRPM8 antagonist.
- Author
-
Gardiner JC, Kirkup AJ, Curry J, Humphreys S, O'Regan P, Postlethwaite M, Young KC, Kitching L, Ethell BT, Winpenny D, and McMurray G
- Subjects
- Anilides pharmacology, Animals, Body Temperature Regulation, Carbamates pharmacology, Female, Ganglia, Spinal metabolism, Guinea Pigs, HEK293 Cells, Humans, Male, Menthol analogs & derivatives, Menthol pharmacology, Muscle, Smooth metabolism, Neurons metabolism, TRPM Cation Channels agonists, TRPM Cation Channels genetics, TRPM Cation Channels physiology, Urethra metabolism, Urinary Bladder metabolism, TRPM Cation Channels antagonists & inhibitors
- Abstract
Patients with overactive bladder often exhibit abnormal bladder contractions in response to intravesical cold saline (positive ice-water test). The molecular entity involved in cold sensation within the urinary bladder is unknown, but a potential candidate is the ion channel, transient receptor potential (melastatin)-8 (TRPM8). The objective of the present study was to investigate the role of TRPM8 in a bladder-cooling reflex evoked in anaesthetised guinea-pigs that is comparable to the positive ice-water test seen in patients. Guinea-pig TRPM8 was cloned from L6 dorsal root ganglia (DRG) and expressed in HEK293 cells. Functional agonist- and cold-induced Ca2+ influx and electrophysiology assays were performed in these cells, and for comparison in HEK293 cells expressing human TRPM8, using a novel TRPM8 antagonist, the S-enantiomer of 1-phenylethyl 4-(benzyloxy)-3-methoxybenzyl (2-aminoethyl) carbamate hydrochloride (PBMC). Potency data from these assays was used to calculate intravenous infusion protocols for targeted plasma concentrations of PBMC in studies on micturition reflexes evoked by intravesical infusion of menthol or cold saline in anaesthetised guinea-pigs. Tissue expression of TRPM8 in guinea-pig bladder, urethra and in dorsal root ganglia neurones traced from the bladder was also investigated. TRPM8 mRNA and protein were detected in L6 dorsal root ganglia, bladder urothelium and smooth muscle. PBMC antagonised in vitro activation of human and guinea-pig TRPM8 and reversed menthol and cold-induced facilitation of the micturition reflex at plasma concentrations consistent with in vitro potencies. The present data suggest that the bladder-cooling reflex in the guinea-pig involves TRPM8. The potential significance of TRPM8 in bladder disease states deserves future investigation., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
118. TRPM7 and its potential therapeutic target for periodontal diseases: beyond its role in the dental pulp repair process.
- Author
-
Xu JL and Xia R
- Subjects
- Humans, Cell Differentiation physiology, Cell Movement physiology, Cell Proliferation physiology, Dental Pulp cytology, Protein Serine-Threonine Kinases physiology, Stem Cells cytology, TRPM Cation Channels physiology
- Published
- 2014
- Full Text
- View/download PDF
119. Regulation and localization of transient receptor potential melastatin 2 in rat uterus.
- Author
-
Ahn C, Yang H, Hong EJ, and Jeung EB
- Subjects
- Animals, Female, Rats, Rats, Sprague-Dawley, Stromal Cells chemistry, Stromal Cells physiology, Uterus cytology, Estrous Cycle physiology, TRPM Cation Channels analysis, TRPM Cation Channels physiology, Uterus chemistry, Uterus physiology
- Abstract
The transient receptor potential channels are membrane-binding proteins that are nonselectively permeable for cations, such as Ca(2+) and Mg(2+), in numerous mammalian cells. The extracellular or intracellular ions play key roles in physiological functions including muscle contraction, cytokine production, insulin release, and apoptosis. Although transient receptor potential melastatin (TRPM) channels are implicated in nonreproductive tissues, the presence of TRPM2 has been reported in endometrium of uterus. To examine whether the expression of TRPM2 gene in uterus is due to gonadal steroid hormones or hormone-independent effect, the uterine TRPM2 gene was monitored in uterus of mature rat during estrous cycle and of immature rat after treatment with gonadal steroid estrogen (E2), progesterone (P4) with/without estrogen receptor antagonist Imperial Chemical Industries (ICI) 182780. We examined real-time polymerase chain reaction, Western blot, and immunohistochemistry to demonstrate the expression and localization of the uterine TRPM2 gene. The level of TRPM2 messenger RNA and protein are dramatically induced at proestrus, then dropped to base line levels at metestrus, and restored its level at diestrus. The results imply that uterine TRPM2 expression levels are regulated by gonadal steroid hormone E2. Moreover, the E2-induced TRPM2 expression is inhibited by cotreatment with ICI 182780 or P4. Furthermore, the immune-reactive TRPM2 is observed in myometrium and stromal cell of endometrium and also showed alterations in TRPM2 expression during estrus cycle. This study suggests that TRPM2 may be involved in calcium absorption or uterine contraction and the latter may be related to implantation or labor by endogenous sex steroid hormones., (© The Author(s) 2014.)
- Published
- 2014
- Full Text
- View/download PDF
120. Response to the 'Letter to the editor by Xu JL'.
- Author
-
Cui L, Xu S, Ma D, and Wu B
- Subjects
- Humans, Cell Differentiation physiology, Cell Movement physiology, Cell Proliferation physiology, Dental Pulp cytology, Protein Serine-Threonine Kinases physiology, Stem Cells cytology, TRPM Cation Channels physiology
- Published
- 2014
- Full Text
- View/download PDF
121. [Pathophysiology of immune cells during the progression of cerebral ischemic injury - involvement of TRPM2-mediated induction of iNOS in microglia/macrophage].
- Author
-
Shirakawa H, Sakimoto S, Nakagawa T, and Kaneko S
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, TRPM Cation Channels genetics, Brain Ischemia physiopathology, Macrophages enzymology, Microglia enzymology, Nitric Oxide Synthase Type II physiology, TRPM Cation Channels physiology
- Published
- 2014
- Full Text
- View/download PDF
122. Mouse nasal epithelial innate immune responses to Pseudomonas aeruginosa quorum-sensing molecules require taste signaling components.
- Author
-
Lee RJ, Chen B, Redding KM, Margolskee RF, and Cohen NA
- Subjects
- Acyl-Butyrolactones pharmacology, Animals, Mice, Mice, Knockout, Nitric Oxide physiology, Phospholipase C beta genetics, Phospholipase C beta physiology, Signal Transduction genetics, TRPM Cation Channels genetics, TRPM Cation Channels physiology, Taste genetics, Transducin genetics, Immunity, Innate immunology, Nasal Mucosa immunology, Pseudomonas aeruginosa chemistry, Quorum Sensing, Signal Transduction drug effects, Taste drug effects
- Abstract
We previously observed that the human bitter taste receptor T2R38 is an important component of upper respiratory innate defense because it detects acyl homoserine lactone (AHL) quorum-sensing molecules secreted by Gram-negative bacteria. T2R38 activation in human sinonasal epithelial cells stimulates calcium and NO signals that increase mucociliary clearance, the major physical respiratory defense against inhaled pathogens. While mice do not have a clear T2R38 ortholog, they do have bitter taste receptors capable of responding to T2R38 agonists, suggesting that T2R-mediated innate immune mechanisms may be conserved in mice. We examined whether AHLs activate calcium and NO signaling in mouse nasal epithelial cells, and utilized pharmacology, as well as cells from knockout mice lacking important components of canonical taste signal transduction pathways, to determine if AHL-stimulated responses require taste signaling molecules. We found that AHLs stimulate calcium-dependent NO production that increases mucociliary clearance and thus likely serves an innate immune role against Gram-negative bacteria. These responses require PLCβ2 and TRPM5 taste signaling components, but not α-gustducin. These data suggest the mouse may be a useful model for further studies of T2R-mediated innate immunity., (© The Author(s) 2013 Reprints and permissions: sagepub.co.uk/journalsPermissions.nav.)
- Published
- 2014
- Full Text
- View/download PDF
123. Transient receptor potential melastatin 2 protects mice against polymicrobial sepsis by enhancing bacterial clearance.
- Author
-
Qian X, Numata T, Zhang K, Li C, Hou J, Mori Y, and Fang X
- Subjects
- Alanine Transaminase blood, Animals, Blotting, Western, Body Burden, Bone Marrow Cells drug effects, Bronchoalveolar Lavage Fluid cytology, Calcium metabolism, Cells, Cultured, Cytokines blood, Heme Oxygenase-1 biosynthesis, Heme Oxygenase-1 genetics, Lung pathology, Macrophages, Peritoneal physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocytes physiology, Organ Size physiology, Patch-Clamp Techniques, Phagocytosis genetics, Phagocytosis physiology, Real-Time Polymerase Chain Reaction, Sepsis mortality, TRPM Cation Channels genetics, Bacteria, Sepsis genetics, Sepsis microbiology, TRPM Cation Channels physiology
- Abstract
Background: Recent studies suggest that the transient receptor potential melastatin 2 (TRPM2) channel plays an important role in inflammation and immune response. However, the role and mechanism of TRPM2 in polymicrobial sepsis remain unclear., Methods: The authors explored the effects of genetic disruption of TRPM2 on mortality (n = 15), bacterial clearance (n = 6), organ injury, and systemic inflammation during cecal ligation and puncture-induced sepsis. Electrophysiology, immunoblot, bacterial clearance experiment, and quantitative real-time polymerase chain reaction were used to explore the role and mechanism of TRPM2 in sepsis., Results: After cecal ligation and puncture, Trpm2-knockout mice had increased mortality compared with wild-type mice (73.3 vs. 40%, P = 0.0289). The increased mortality was associated with increased bacterial burden, organ injury, and systemic inflammation. TRPM2-mediated Ca influx plays an important role in lipopolysaccharide or cecal ligation and puncture-induced heme oxygenase-1 (HO-1) expression in macrophage. HO-1 up-regulation decreased bacterial burden both in wild-type bone marrow-derived macrophages and in cecal ligation and puncture-induced septic wild-type mice. Disruption of TRPM2 decreased HO-1 expression and increased bacterial burden in bone marrow-derived macrophages. Pretreatment of Trpm2-knockout bone marrow-derived macrophages with HO-1 inducer markedly increased HO-1 expression and decreased bacterial burden. Pretreatment of Trpm2-knockout mice with HO-1 inducer reversed the susceptibility of Trpm2-knockout mice to sepsis by enhancing the bacterial clearance. In addition, septic patients with lower monocytic TRPM2 and HO-1 messenger RNA levels had a worse outcome compared with septic patients with normal monocytic TRPM2 and HO-1 messenger RNA levels. TRPM2 levels correlated with HO-1 levels in septic patients (r = 0.675, P = 0.001)., Conclusion: The study data demonstrate a protective role of TRPM2 in controlling bacterial clearance during polymicrobial sepsis possibly by regulating HO-1 expression.
- Published
- 2014
- Full Text
- View/download PDF
124. TRPM7 involvement in cancer: a potential prognostic factor.
- Author
-
Dhennin-Duthille I, Gautier M, Korichneva I, and Ouadid-Ahidouch H
- Subjects
- Animals, Apoptosis physiology, Biomarkers, Tumor, Calcium metabolism, Cell Differentiation physiology, Cell Division physiology, Cell Movement physiology, Embryonic Development physiology, Homeostasis, Humans, Ion Transport, Mammals metabolism, Neoplasm Proteins chemistry, Neoplasms chemistry, Neoplasms mortality, Prognosis, Protein Processing, Post-Translational physiology, Protein Serine-Threonine Kinases chemistry, Protein Structure, Tertiary, Signal Transduction physiology, TRPM Cation Channels chemistry, Magnesium metabolism, Neoplasm Proteins physiology, Protein Serine-Threonine Kinases physiology, TRPM Cation Channels physiology
- Abstract
Calcium (Ca(2+)) and magnesium (Mg(2+)) are important metal elements that regulate a variety of cellular processes such as proliferation, migration, and apoptosis, in cancer cells. Among the ionic channels mediating intracellular entry, the transient receptor potential melastatin type 7 (TRPM7) channel is of particular interest, it being a non-selective, cationic channel mediating both Ca(2+) and Mg(2+) influx. TRPM7 is highly expressed in a number of human cancer tissues and cell lines. In this review, we summarise current knowledge on the physiological role of the dual function TRPM7 chanzyme, the potential application of TRPM7 as a diagnostic and prognostic marker of cancer progression with respect to clinical and pathological characteristics, and the molecular mechanisms implicated in cancerogenesis that specifically involve Ca(2+) and Mg(2+) influx through TRPM7 or kinase activity and interaction with cytoskeletal proteins.
- Published
- 2014
- Full Text
- View/download PDF
125. The effect of TRPM7 suppression on the proliferation, migration and osteogenic differentiation of human dental pulp stem cells.
- Author
-
Cui L, Xu SM, Ma DD, and Wu BL
- Subjects
- Cells, Cultured, Humans, Cell Differentiation physiology, Cell Movement physiology, Cell Proliferation physiology, Dental Pulp cytology, Protein Serine-Threonine Kinases physiology, Stem Cells cytology, TRPM Cation Channels physiology
- Abstract
Aim: To investigate the role of the Ca(2+) -Mg(2+) ion channel TRPM7 in the proliferation, migration and osteogenic differentiation of human dental pulp stem cells (hDPSCs)., Methodology: Immunohistochemistry was used to localize expression of TRPM7 in human dental pulp tissues and in cultured hDPSCs. Isolated hDPSCs were infected with recombinant lentiviruses expressing short hairpin RNA (shRNA) specific for TRPM7, or control shRNA, in order to suppress TRPM7 mRNA expression and investigate its functional role. The proliferation of the shRNA-infected hDPSCs was evaluated using both an MTT assay to measure viable cell numbers and cell cycle analysis. Cell migration was assessed using a transwell assay. The dynamic mRNA expression of TRPM7 during osteogenic differentiation of hDPSCs and the effect of shRNA specific for TRPM7 on hDPSC osteogenic differentiation were evaluated by real-time PCR., Results: TRPM7 expression was widespread in human dental pulp tissue and was detected mainly in the cytomembrane and cytoplasm of hDPSCs. Suppression of TRPM7 inhibited both the proliferation and the migratory capacity of hDPSCs. TRPM7 mRNA expression was elevated during osteogenic differentiation of hDPSCs. TRPM7-specific shRNA inhibited osteogenic differentiation of hDPSCs, with downregulated mRNA expression of the osteogenic markers alkaline phosphatase (ALP), dentine sialophosphoprotein (DSPP), bone sialoprotein (BSP), runt-related transcription factor (RUNX2) and osterix (OSX)., Conclusions: TRPM7 was involved in the regulation of hDPSC proliferation, migration and osteogenic differentiation and may play a role in the dental pulp repair process., (© 2013 International Endodontic Journal. Published by John Wiley & Sons Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
126. TRPM4 channels in the cardiovascular system.
- Author
-
Kruse M and Pongs O
- Subjects
- Animals, Cardiovascular System metabolism, Humans, TRPM Cation Channels genetics, TRPM Cation Channels metabolism, Cardiovascular Physiological Phenomena, TRPM Cation Channels physiology
- Abstract
The non-selective Transient Receptor Potential Melastatin 4 (TRPM4) cation channel is abundantly expressed in cardiac cells, being involved in several aspects of cardiac rhythmicity, including cardiac conduction, pace making and action-potential repolarization. Dominantly inherited mutations in the TRPM4 gene are associated with the cardiac bundle-branch disorder progressive familial heart block type I (PFHBI) and isolated cardiac conduction disease (ICCD) giving rise to atrio-ventricular conduction block (AVB), right bundle branch block, bradycardia, and the Brugada syndrome. The mutant phenotypes closely resemble those associated with mutations in the SCN5A gene, encoding the voltage-gated Na(+) channel NaV1.5. These observations and the unexpected partnership with sulfonylurea-receptors (SURs) makes the TRPM4 channel a promising novel target for treatment of cardiac disorders., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
127. Involvement of TRPM2 and L-type Ca²⁺ channels in Ca²⁺ entry and cell death induced by hydrogen peroxide in rat β-cell line RIN-5F.
- Author
-
Ishii M, Hagiwara T, Mori Y, and Shimizu S
- Subjects
- Animals, Calcium Channel Blockers pharmacology, Calcium Channels, L-Type drug effects, Cell Death drug effects, Cell Death genetics, Cell Line, Tumor, Enzyme Inhibitors pharmacology, HEK293 Cells, Humans, Insulinoma pathology, Poly(ADP-ribose) Polymerase Inhibitors, Rats, TRPM Cation Channels metabolism, Calcium metabolism, Calcium Channels, L-Type physiology, Hydrogen Peroxide adverse effects, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells pathology, Insulinoma metabolism, TRPM Cation Channels physiology
- Abstract
Ca²⁺ overload is one of the mechanisms for H₂O₂-induced cell death in rat pancreatic β-cell line RIN-5F cells. RIN-5F cells express TRPM2, which is a Ca²⁺-permeable channel activated by H₂O₂, and voltage-dependent L-type Ca²⁺ channels, both of which induce Ca²⁺ entry by H₂O₂. This study examined the contribution of these channels to H₂O₂-induced Ca²⁺ entry and cell death in RIN-5F cells. Cytosolic Ca²⁺ concentration was measured using fura-2 as a Ca²⁺ indicator. Cell death was estimated by trypan blue exclusion. Pre-treatment with poly(ADP-ribose) polymerase (PARP) inhibitors, which inhibit TRPM2 activation, strongly reduced Ca²⁺ entry by H₂O₂. The PARP inhibitors used had no effect on the Ca²⁺ elevation by voltage-dependent L-type Ca²⁺ channels. On the other hand, pre-treatment with L-type Ca²⁺ channel blockers, which did not affect TRPM2 activation, partly reduced H₂O₂-induced Ca²⁺ entry. Treatment with PARP inhibitors but not L-type Ca²⁺ channel blockers, around the early phase in H₂O₂-induced Ca²⁺ elevation, also reduced the late phase. Moreover, H₂O₂-induced RIN-5F cell death was strongly attenuated by PARP inhibitors, in compared to L-type Ca²⁺ channel blockers. Our results suggest that TRPM2 channels rather than L-type Ca²⁺ channels primarily contribute to H₂O₂-induced Ca²⁺ entry and cell death.
- Published
- 2014
- Full Text
- View/download PDF
128. TRPM2: a multifunctional ion channel for oxidative stress sensing.
- Author
-
Ru X and Yao X
- Subjects
- Adenosine Diphosphate Ribose metabolism, Calcium physiology, Calcium Channels physiology, Humans, Hydrogen Peroxide metabolism, Reactive Oxygen Species metabolism, Oxidative Stress, TRPM Cation Channels physiology
- Abstract
Transient receptor potential (TRP) superfamily is a superfamily of cation channels that can be divided into seven subfamilies. TRPM2 is the second member of the TRPM subfamily, which includes eight members, namely TRPM1-8. TRPM2 is widely expressed in excitable and non-excitable cells, where it forms a Ca(2+)-permeable cation channel and performs diverse cellular functions. TRPM2 channels are activated by ADP-ribose (ADPR), Ca(2+), H2O2 and other reactive oxygen species (ROS). It is established that TRPM2 serves as a cellular sensor for oxidative stress, mediating oxidative stress-induced [Ca(2+)]i increase and contributing to pathological processes in many cell types. Accumulating evidence has indicated that TRPM2 is a potential therapeutic target for oxidative stress-related diseases. This review will highlight recent progress in this field.
- Published
- 2014
129. Structural requirements of steroidal agonists of transient receptor potential melastatin 3 (TRPM3) cation channels.
- Author
-
Drews A, Mohr F, Rizun O, Wagner TF, Dembla S, Rudolph S, Lambert S, Konrad M, Philipp SE, Behrendt M, Marchais-Oberwinkler S, Covey DF, and Oberwinkler J
- Subjects
- Animals, Base Sequence, Binding Sites, HEK293 Cells, Humans, Mice, Molecular Sequence Data, Nifedipine pharmacology, Pregnenolone pharmacology, TRPM Cation Channels agonists, TRPM Cation Channels chemistry, TRPM Cation Channels metabolism, TRPM Cation Channels physiology
- Abstract
Background and Purpose: Transient receptor potential melastatin 3 (TRPM3) proteins form non-selective but calcium-permeable membrane channels, rapidly activated by extracellular application of the steroid pregnenolone sulphate and the dihydropyridine nifedipine. Our aim was to characterize the steroid binding site by analysing the structural chemical requirements for TRPM3 activation., Experimental Approach: Whole-cell patch-clamp recordings and measurements of intracellular calcium concentrations were performed on HEK293 cells transfected with TRPM3 (or untransfected controls) during superfusion with pharmacological substances., Key Results: Pregnenolone sulphate and nifedipine activated TRPM3 channels supra-additively over a wide concentration range. Other dihydropyridines inhibited TRPM3 channels. The natural enantiomer of pregnenolone sulphate was more efficient in activating TRPM3 channels than its synthetic mirror image. However, both enantiomers exerted very similar inhibitory effects on proton-activated outwardly rectifying anion channels. Epiallopregnanolone sulphate activated TRPM3 almost equally as well as pregnenolone sulphate. Exchanging the sulphate for other chemical moieties showed that a negative charge at this position is required for activating TRPM3 channels., Conclusions and Implications: Our data demonstrate that nifedipine and pregnenolone sulphate act at different binding sites when activating TRPM3. The latter activates TRPM3 by binding to a chiral and thus proteinaceous binding site, as inferred from the differential effects of the enantiomers. The double bond between position C5 and C6 of pregnenolone sulphate is not strictly necessary for the activation of TRPM3 channels, but a negative charge at position C3 of the steroid is highly important. These results provide a solid basis for understanding mechanistically the rapid chemical activation of TRPM3 channels., (© 2013 The Authors. British Journal of Pharmacology published by John Wiley &. Sons Ltd on behalf of The British Pharmacological Society.)
- Published
- 2014
- Full Text
- View/download PDF
130. Rhinovirus upregulates transient receptor potential channels in a human neuronal cell line: implications for respiratory virus-induced cough reflex sensitivity.
- Author
-
Abdullah H, Heaney LG, Cosby SL, and McGarvey LP
- Subjects
- Calcium Channels physiology, Cell Line, Cough virology, Flow Cytometry, Humans, Nerve Tissue Proteins physiology, Neuroblastoma, Picornaviridae Infections, Respiratory Tract Infections physiopathology, TRPA1 Cation Channel, TRPM Cation Channels physiology, TRPV Cation Channels physiology, Tumor Cells, Cultured, Up-Regulation physiology, Virus Replication physiology, Cough physiopathology, Respiratory Tract Infections virology, Rhinovirus physiology, Transient Receptor Potential Channels physiology, Virus Diseases physiopathology
- Abstract
Background: The mechanism underlying respiratory virus-induced cough hypersensitivity is unknown. Upregulation of airway neuronal receptors responsible for sensing physical and chemical stimuli is one possibility, and the transient receptor potential (TRP) channel family are potential candidates. We have used an in vitro model of sensory neurons and human rhinovirus (HRV-16) to study the effect of virus infection on TRP expression., Methods: IMR-32 neuroblastoma cells were differentiated in culture to express three TRP channels: TRPV1, TRPA1 and TRPM8. Flow cytometry and qRT-PCR were used to measure TRP channel protein and mRNA levels following inoculation with live virus, inactivated virus, virus-induced soluble factors or pelleted virus particles. Multiplex bioassay was used to determine nerve growth factor (NGF), interleukin (IL)-1β, IL-6 and IL-8 levels in response to infection., Results: Early upregulation of TRPA1 and TRPV1 expression occurred 2-4 h post infection. This was independent of replicating virus as virus-induced soluble factors alone were sufficient to increase channel expression 50-fold and 15-fold, respectively. NGF, IL-6 and IL-8 levels, increased in infected cell supernatants, represent possible candidates. In contrast, TRPM8 expression was maximal at 48 h (9.6-fold) and required virus replication rather than soluble factors., Conclusions: We show for the first time that rhinovirus can infect neuronal cells. Furthermore, infection causes upregulation of TRP channels by channel-specific mechanisms. The increase in TRPA1 and TRPV1 levels can be mediated by soluble factors induced by infection whereas TRPM8 requires replicating virus. TRP channels may be novel therapeutic targets for controlling virus-induced cough.
- Published
- 2014
- Full Text
- View/download PDF
131. Transient receptor potential melastatin 7 (TRPM7) contributes to H2O2-induced cardiac fibrosis via mediating Ca(2+) influx and extracellular signal-regulated kinase 1/2 (ERK1/2) activation in cardiac fibroblasts.
- Author
-
Guo JL, Yu Y, Jia YY, Ma YZ, Zhang BY, Liu PQ, Chen SR, and Jiang JM
- Subjects
- Actins metabolism, Animals, Cells, Cultured, Collagen Type I metabolism, Connective Tissue Growth Factor metabolism, Fibronectins metabolism, Fibrosis, Male, Myocardium metabolism, Phosphorylation, RNA Interference, RNA, Small Interfering, Rats, Sprague-Dawley, TRPM Cation Channels genetics, Transforming Growth Factor beta metabolism, Calcium metabolism, Hydrogen Peroxide adverse effects, MAP Kinase Signaling System genetics, MAP Kinase Signaling System physiology, Mitogen-Activated Protein Kinase 3 metabolism, Myocardium pathology, TRPM Cation Channels physiology
- Abstract
Transient receptor potential melastatin 7 (TRPM7), a Ca(2+)-nonselective cation channel, plays a key role in the pathophysiological response of multiple cell types. However, the role of TRPM7 channels in hydrogen peroxide (H2O2)-induced cardiac fibrosis remains unclear. This study aimed to explore whether TRPM7 channels are involved in H2O2-induced cardiac fibrosis and the underlying mechanisms. Our results showed that 2-aminoethoxydiphenylborate (2-APB), which is commonly used to block TRPM7 channels, inhibited H2O2-induced cardiac fibrosis via attenuating the overexpression of important fibrogenic biomarkers and growth factors in cardiac fibroblasts, including collagen type I (Col I), fibronectin (FN), smooth muscle α-actin (α-SMA), connective tissue growth factor (CTGF), and transforming growth factor-β1 (TGF-β1). In addition, 2-APB also decreased H2O2-mediated elevation of the concentration of intracellular Ca(2+) ([Ca(2+)]i). Meanwhile, silencing TRPM7 channels by shRNA interference also impaired the increased [Ca(2+)]i and upregulation of Col I, FN, α-SMA, CTGF, and TGF-β1 induced by H2O2. Furthermore, we found that H2O2-mediated activation of extracellular signal-regulated kinase 1/2 (ERK1/2) decreased in TRPM7-shRNA cells and Ca(2+)-free culture media. These results demonstrated that TRPM7 channels contributed to H2O2-induced cardiac fibrosis and suggested that this contribution may be through mediating Ca(2+) influx and phosphorylation of ERK1/2.
- Published
- 2014
- Full Text
- View/download PDF
132. Phosphoinositide regulation of TRP channels.
- Author
-
Rohacs T
- Subjects
- Animals, Humans, Signal Transduction physiology, TRPC Cation Channels physiology, TRPM Cation Channels physiology, TRPV Cation Channels physiology, Phosphatidylinositols physiology, Transient Receptor Potential Channels physiology
- Abstract
Transient Receptor Potential (TRP) channels are activated by stimuli as diverse as heat, cold, noxious chemicals, mechanical forces, hormones, neurotransmitters, spices, and voltage. Besides their presumably similar general architecture, probably the only common factor regulating them is phosphoinositides. The regulation of TRP channels by phosphoinositides is complex. There are a large number of TRP channels where phosphatidylinositol 4,5 bisphosphate [PI(4,5)P2 or PIP2] acts as a positive cofactor, similarly to many other ion channels. In several cases, however, PI(4,5)P2 inhibits TRP channel activity, sometimes even concurrently with the activating effect. This chapter will provide a comprehensive overview of the literature on regulation of TRP channels by membrane phosphoinositides.
- Published
- 2014
- Full Text
- View/download PDF
133. The interaction of transient receptor potential melastatin 7 with macrophages promotes vascular adventitial remodeling in transverse aortic constriction rats.
- Author
-
Li Y, Jiang H, Ruan C, Zhong J, Gao P, Zhu D, Niu W, and Guo S
- Subjects
- Animals, Annexins metabolism, Aorta pathology, Blotting, Western, Carotid Artery, Common pathology, Cells, Cultured, Constriction, Pathologic, Fibroblasts pathology, Hypertension physiopathology, Male, Myofibroblasts pathology, RNA, Small Interfering genetics, Rats, Rats, Sprague-Dawley, TRPM Cation Channels antagonists & inhibitors, TRPM Cation Channels genetics, Transfection, Up-Regulation drug effects, Vasculitis pathology, Aorta physiology, Macrophages physiology, TRPM Cation Channels physiology
- Abstract
Transient receptor potential melastatin 7 (TRPM7), a novel channel kinase, has been recently identified in the vasculature. However, its regulation and function in vascular diseases remain poorly understood. To address this lack of knowledge, we sought to examine whether TRPM7 can mediate the vascular remodeling process induced by pressure overload in the right common carotid artery proximal to the band (RCCA-B) in male Sprague-Dawley rats with transverse aortic constriction (TAC). The contribution of TRPM7 to amplified vascular remodeling after TAC was tested using morphometric and western blot analyses. Pressure overload-induced vascular wall thickening, especially in the adventitia, was readily detected in RCCA-B. The TRPM7 level was increased with a simultaneous accumulation of macrophages in the adventitia of RCCA-B, whereas the anti-inflammatory molecule annexin-1, a TRPM7 downstream target, was decreased. After the addition of the TRPM7 inhibitor 2-aminoethoxydiphenyl borate (2-APB), significant reductions in macrophage accumulation as well as the expression of monocyte chemotactic protein-1, SM-22-α and collagen I were observed, whereas annexin-1 was rescued. Finally, in cultured vascular adventitial fibroblasts treated with macrophage-conditioned medium, there were marked increases in the expression of TRPM7 and SM-22-α with a concurrent reduction in annexin-1 expression; these effects were largely prevented by treatment with 2-APB and specific anti-TRPM7 small interfering RNA. Our findings provide the first demonstration of the potential regulatory roles of TRPM7 in the vascular inflammation, pressure overload-mediated vascular adventitial collagen accumulation and cell phenotypic transformation in TAC rats. The targeting of TRPM7 has potential therapeutic importance for vascular diseases.
- Published
- 2014
- Full Text
- View/download PDF
134. Effect of glibenclamide on the prevention of secondary brain injury following ischemic stroke in humans.
- Author
-
Khanna A, Walcott BP, Kahle KT, and Simard JM
- Subjects
- Brain Diseases etiology, Brain Edema etiology, Brain Edema prevention & control, Clinical Trials, Phase II as Topic, Humans, Intracranial Hemorrhages etiology, Intracranial Hemorrhages prevention & control, Sulfonylurea Receptors genetics, Sulfonylurea Receptors physiology, TRPM Cation Channels genetics, TRPM Cation Channels physiology, Brain Diseases prevention & control, Brain Ischemia complications, Brain Ischemia drug therapy, Glyburide therapeutic use, Stroke complications, Stroke drug therapy
- Abstract
Cerebral edema and hemorrhagic conversion are common, potentially devastating complications of ischemic stroke and are associated with high rates of mortality and poor functional outcomes. Recent work exploring the molecular pathophysiology of the neurogliovascular unit in ischemic stroke suggests that deranged cellular ion homeostasis due to altered function and regulation of ion pumps, channels, and secondary active transporters plays an integral role in the development of cytotoxic and vasogenic edema and hemorrhagic conversion. Among these proteins involved in ion homeostasis, the ischemia-induced, nonselective cation conductance formed by the SUR1-TRPM4 protein complex appears to play a prominent role and is potently inhibited by glibenclamide, an FDA-approved drug commonly used in patients with Type 2 diabetes. Several robust preclinical studies have demonstrated the efficacy of glibenclamide blockade of SUR1-TRPM4 activity in reducing edema and hemorrhagic conversion in rodent models of ischemic stroke, prompting the study of the potential protective effects of glibenclamide in humans in an ongoing prospective phase II clinical trial. Preliminary data suggest glibenclamide significantly reduces cerebral edema and lowers the rate of hemorrhagic conversion following ischemic stroke, suggesting the potential use of glibenclamide to improve outcomes in humans.
- Published
- 2014
- Full Text
- View/download PDF
135. Selective inhibition of KCa3.1 channels mediates adenosine regulation of the motility of human T cells.
- Author
-
Chimote AA, Hajdu P, Kucher V, Boiko N, Kuras Z, Szilagyi O, Yun YH, and Conforti L
- Subjects
- 8-Bromo Cyclic Adenosine Monophosphate pharmacology, Adenosine analogs & derivatives, Calcium physiology, Calcium Channel Blockers pharmacology, Cell Movement drug effects, Cells, Cultured, Cyclic AMP-Dependent Protein Kinase Type I antagonists & inhibitors, Cyclic AMP-Dependent Protein Kinase Type I physiology, Female, Humans, Immunologic Surveillance physiology, Intercellular Adhesion Molecule-1, Interleukin-2 metabolism, Intermediate-Conductance Calcium-Activated Potassium Channels physiology, Ion Transport drug effects, Kv1.3 Potassium Channel physiology, Lymphocyte Activation, Male, Patch-Clamp Techniques, Phenethylamines pharmacology, Protein Serine-Threonine Kinases, Pyrazoles pharmacology, Pyrimidines pharmacology, Receptor, Adenosine A2A physiology, T-Lymphocytes cytology, T-Lymphocytes metabolism, TRPM Cation Channels physiology, Triazoles pharmacology, Adenosine pharmacology, Intermediate-Conductance Calcium-Activated Potassium Channels antagonists & inhibitors, T-Lymphocytes drug effects
- Abstract
Adenosine, a purine nucleoside, is present at high concentrations in tumors, where it contributes to the failure of immune cells to eliminate cancer cells. The mechanisms responsible for the immunosuppressive properties of adenosine are not fully understood. We tested the hypothesis that adenosine's immunosuppressive functions in human T lymphocytes are in part mediated via modulation of ion channels. The activity of T lymphocytes relies on ion channels. KCa3.1 and Kv1.3 channels control cytokine release and, together with TRPM7, regulate T cell motility. Adenosine selectively inhibited KCa3.1, but not Kv1.3 and TRPM7, in activated human T cells. This effect of adenosine was mainly mediated by A2A receptors, as KCa3.1 inhibition was reversed by SCH58261 (selective A2A receptor antagonist), but not by MRS1754 (A2B receptor antagonist), and it was mimicked by the A2A receptor agonist CGS21680. Furthermore, it was mediated by the cAMP/protein kinase A isoform (PKAI) signaling pathway, as adenylyl-cyclase and PKAI inhibition prevented adenosine effect on KCa3.1. The functional implication of the effect of adenosine on KCa3.1 was determined by measuring T cell motility on ICAM-1 surfaces. Adenosine and CGS21680 inhibited T cell migration. Comparable effects were obtained by KCa3.1 blockade with TRAM-34. Furthermore, the effect of adenosine on cell migration was abolished by pre-exposure to TRAM-34. Additionally, adenosine suppresses IL-2 secretion via KCa3.1 inhibition. Our data indicate that adenosine inhibits KCa3.1 in human T cells via A2A receptor and PKAI, thereby resulting in decreased T cell motility and cytokine release. This mechanism is likely to contribute to decreased immune surveillance in solid tumors.
- Published
- 2013
- Full Text
- View/download PDF
136. Flavanones that selectively inhibit TRPM3 attenuate thermal nociception in vivo.
- Author
-
Straub I, Krügel U, Mohr F, Teichert J, Rizun O, Konrad M, Oberwinkler J, and Schaefer M
- Subjects
- Animals, Calcium metabolism, Cell Survival drug effects, Dose-Response Relationship, Drug, Female, Flavonoids pharmacology, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Pregnenolone pharmacology, Rats, Rats, Wistar, TRPM Cation Channels physiology, TRPV Cation Channels antagonists & inhibitors, Flavanones pharmacology, Hyperalgesia drug therapy, TRPM Cation Channels antagonists & inhibitors
- Abstract
Transient receptor potential melastatin 3 (TRPM3) is a calcium-permeable nonselective cation channel that is expressed in a subset of dorsal root (DRG) and trigeminal ganglia sensory neurons. TRPM3 can be activated by the neurosteroid pregnenolone sulfate (PregS) and heat. TRPM3⁻/⁻ mice display an impaired sensation of noxious heat and thermal hyperalgesia. We have previously shown that TRPM3 is blocked by the citrus fruit flavanones hesperetin, naringenin, and eriodictyol as well as by ononetin, a deoxybenzoin from Ononis spinosa. To further improve the tolerability, potency, and selectivity of TRPM3 blockers, we conducted a hit optimization procedure by rescreening a focused library that was composed of chemically related compounds. Within newly identified TRPM3 blockers, isosakuranetin and liquiritigenin displayed favorable properties with respect to their inhibitory potency and a selective mode of action. Isosakuranetin, a flavanone whose glycoside is contained in blood oranges and grapefruits, displayed an IC₅₀ of 50 nM and is to our knowledge the most potent inhibitor of TRPM3 identified so far. Both compounds exhibited a marked specificity for TRPM3 compared with other sensory TRP channels, and blocked PregS-induced intracellular free Ca²⁺ concentration signals and ionic currents in freshly isolated DRG neurons. Furthermore, isosakuranetin and previously identified hesperetin significantly reduced the sensitivity of mice to noxious heat and PregS-induced chemical pain. Because the physiologic functions of TRPM3 channels are still poorly defined, the development and validation of potent and selective blockers is expected to contribute to clarifying the role of TRPM3 in vivo.
- Published
- 2013
- Full Text
- View/download PDF
137. TRPM7 channel regulates PDGF-BB-induced proliferation of hepatic stellate cells via PI3K and ERK pathways.
- Author
-
Fang L, Zhan S, Huang C, Cheng X, Lv X, Si H, and Li J
- Subjects
- Animals, Becaplermin, Cells, Cultured, Collagen Type I, alpha 1 Chain, Fibroblasts enzymology, Fibroblasts metabolism, Fibroblasts pathology, Hepatic Stellate Cells drug effects, Hepatic Stellate Cells pathology, Liver Cirrhosis metabolism, Liver Cirrhosis pathology, MAP Kinase Signaling System drug effects, Male, Rats, Rats, Sprague-Dawley, Cell Proliferation drug effects, Hepatic Stellate Cells enzymology, MAP Kinase Signaling System physiology, Phosphatidylinositol 3-Kinases physiology, Proto-Oncogene Proteins c-sis physiology, TRPM Cation Channels physiology
- Abstract
TRPM7, a non-selective cation channel of the TRP channel superfamily, is implicated in diverse physiological and pathological processes including cell proliferation. Recently, TRPM7 has been reported in hepatic stellate cells (HSCs). Here, we investigated the contribution role of TRPM7 in activated HSC-T6 cell (a rat hepatic stellate cell line) proliferation. TRPM7 mRNA and protein were measured by RT-PCR and Western blot in rat model of liver fibrosis in vivo and PDGF-BB-activated HSC-T6 cells in vitro. Both mRNA and protein of TRPM7 were dramatically increased in CCl4-treated rat livers. Stimulation of HSC-T6 cells with PDGF-BB resulted in a time-dependent increase of TRPM7 mRNA and protein. However, PDGF-BB-induced HSC-T6 cell proliferation was inhibited by non-specific TRPM7 blocker 2-aminoethoxydiphenyl borate (2-APB) or synthetic siRNA targeting TRPM7, and this was accompanied by downregulation of cell cycle proteins, cyclin D1, PCNA and CDK4. Blockade of TRPM7 channels also attenuated PDGF-BB induced expression of myofibroblast markers as measured by the induction of α-SMA and Col1α1. Furthermore, the phosphorylation of ERK and AKT, associated with cell proliferation, decreased in TRPM7 deficient HSC-T6 cells. These observations suggested that TRPM7 channels contribute to perpetuated fibroblast activation and proliferation of PDGF-BB induced HSC-T6 cells via the activation of ERK and PI3K pathways. Therefore, TRPM7 may constitute a useful target for the treatment of liver fibrosis., (© 2013.)
- Published
- 2013
- Full Text
- View/download PDF
138. [Involvement of spinally-infiltrated immune cells in peripheral nerve injury-induced neuropathic pain: roles of TRPM2].
- Author
-
Nakagawa T, Isami K, Haraguchi K, So K, Asakura K, Shirakawa H, and Kaneko S
- Subjects
- Animals, Humans, Mice, Reactive Oxygen Species, Macrophages immunology, Macrophages pathology, Neuralgia immunology, Neuralgia pathology, Neuroimmunomodulation, Peripheral Nerve Injuries immunology, Peripheral Nerve Injuries pathology, Spinal Cord immunology, Spinal Cord pathology, T-Lymphocytes immunology, T-Lymphocytes pathology, TRPM Cation Channels physiology
- Published
- 2013
- Full Text
- View/download PDF
139. Control of urinary bladder smooth muscle excitability by the TRPM4 channel modulator 9-phenanthrol.
- Author
-
Parajuli SP, Hristov KL, Sullivan MN, Xin W, Smith AC, Earley S, Malysz J, and Petkov GV
- Subjects
- Animals, Male, Muscle, Smooth physiology, Myocytes, Smooth Muscle physiology, TRPM Cation Channels physiology, Urinary Bladder physiology
- Abstract
The Ca (2+)-activated monovalent cation selective transient receptor potential melastatin 4 (TRPM4) channel has been recently identified in detrusor smooth muscle (DSM) of the urinary bladder. Two recent publications by our research group provide evidence in support of the novel hypothesis that TRPM4 channels enhance DSM excitability and contractility. This is a critical question as prior studies have primarily targeted hyperpolarizing currents facilitated by K(+) channels, but the depolarizing component in DSM cells is not well understood. For the first time, we utilized the selective TRPM4 channel inhibitor, 9-phenanthrol, to investigate TRPM4 channel functional effects in DSM at both cellular and tissue levels in rodents. Our new data presented here showed that in rat DSM cells, 9-phenanthrol attenuates spontaneous inward currents in the presence of the muscarinic receptor agonist, carbachol, thus reducing DSM cell excitability. In support of our original hypothesis, we found that TRPM4 channel mRNA levels are much higher in DSM vs. vascular smooth muscle and that inhibition of TRPM4 channels can potentially attenuate DSM excitability. Thus, we postulate the novel concept that selective pharmacological inhibition of TRPM4 channels can limit both excitability and contractility of DSM.
- Published
- 2013
- Full Text
- View/download PDF
140. Transient receptor potential melastatin 7 is involved in oestrogen receptor-negative metastatic breast cancer cells migration through its kinase domain.
- Author
-
Guilbert A, Gautier M, Dhennin-Duthille I, Rybarczyk P, Sahni J, Sevestre H, Scharenberg AM, and Ouadid-Ahidouch H
- Subjects
- Calcium metabolism, Cell Movement drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Neoplasm Metastasis, Phosphotransferases chemistry, Phosphotransferases physiology, Protein Structure, Tertiary physiology, RNA, Small Interfering pharmacology, Receptors, Estrogen metabolism, TRPM Cation Channels antagonists & inhibitors, TRPM Cation Channels chemistry, Tumor Cells, Cultured, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Movement genetics, TRPM Cation Channels physiology
- Abstract
Oestrogen receptor negative (ER(-)) invasive ductal carcinoma (IDC) represents a significant clinical challenge and therefore prompts the discovery of novel biomarkers. Transient receptor potential melastatin 7 (TRPM7), a channel protein that also contains a regulatory kinase domain, is overexpressed in IDC and regulates migration. However, the molecular mechanism remains poorly defined. Here, we examined whether TRPM7 regulates migration by its channel function or by its kinase domain. A Magnesium Inhibited Cation current was recorded in two ER(-) highly metastatic breast cancer cell lines. Down-regulation of TRPM7 neither affected Ca(2+)-, nor Mg(2+)-homoeostasis but significantly reduced cell migration via a Ca(2+)-independent pathway. Notably, the overexpression of the truncated kinase domain form of TRPM7 decreased cell migration, while the overexpression of the wild-type form strongly increased it. Concomitantly, TRPM7 silencing reduced the myosin IIA heavy chain phosphorylation. Furthermore, we found higher TRPM7 expression in ER(-) IDC tissues and lymph nodes than in the non-invasive tumoural samples. In conclusion, TRPM7 plays a critical role in breast cancer cell migration through its kinase domain, and our data support the consideration of using TRPM7 as a novel biomarker and a potential therapeutic target in the treatment of human ER(-) IDC., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
141. Modulation of NMDAR subunit expression by TRPM2 channels regulates neuronal vulnerability to ischemic cell death.
- Author
-
Alim I, Teves L, Li R, Mori Y, and Tymianski M
- Subjects
- Animals, Brain Ischemia pathology, Cell Death physiology, Cell Survival physiology, Cells, Cultured, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neural Pathways physiology, Neurons pathology, Protein Subunits biosynthesis, Receptors, N-Methyl-D-Aspartate biosynthesis, Brain Ischemia metabolism, Brain Ischemia prevention & control, Down-Regulation genetics, Neurons metabolism, Protein Subunits genetics, Receptors, N-Methyl-D-Aspartate genetics, TRPM Cation Channels physiology
- Abstract
Neuronal vulnerability to ischemia is dependent on the balance between prosurvival and prodeath cellular signaling. In the latter, it is increasingly appreciated that toxic Ca(2+) influx can occur not only via postsynaptic glutamate receptors, but also through other cation conductances. One such conductance, the Transient receptor potential melastatin type-2 (TRPM2) channel, is a nonspecific cation channel having homology to TRPM7, a conductance reported to play a key role in anoxic neuronal death. The role of TRPM2 conductances in ischemic Ca(2+) influx has been difficult to study because of the lack of specific modulators. Here we used TRPM2-null mice (TRPM2(-/-)) to study how TRPM2 may modulate neuronal vulnerability to ischemia. TRPM2(-/-) mice subjected to transient middle cerebral artery occlusion exhibited smaller infarcts when compared with wild-type animals, suggesting that the absence of TRPM2 is neuroprotective. Surprisingly, field potentials (fEPSPs) recorded during redox modulation in brain slices taken from TRPM2(-/-) mice revealed increased excitability, a phenomenon normally associated with ischemic vulnerability, whereas wild-type fEPSPs were unaffected. The upregulation in fEPSP in TRPM2(-/-) neurons was blocked selectively by a GluN2A antagonist. This increase in excitability of TRPM2(-/-) fEPSPs during redox modulation depended on the upregulation and downregulation of GluN2A- and GluN2B-containing NMDARs, respectively, and on augmented prosurvival signaling via Akt and ERK pathways culminating in the inhibition of the proapoptotic factor GSK3β. Our results suggest that TRPM2 plays a role in downregulating prosurvival signals in central neurons and that TRPM2 channels may comprise a therapeutic target for preventing ischemic damage.
- Published
- 2013
- Full Text
- View/download PDF
142. TRPM8 is the principal mediator of menthol-induced analgesia of acute and inflammatory pain.
- Author
-
Liu B, Fan L, Balakrishna S, Sui A, Morris JB, and Jordt SE
- Subjects
- Acute Pain physiopathology, Animals, Cells, Cultured, Inflammation drug therapy, Inflammation physiopathology, Male, Menthol pharmacology, Mice, Mice, Inbred C57BL, Mice, Knockout, Pain Measurement drug effects, Acute Pain drug therapy, Analgesia methods, Menthol therapeutic use, Pain Measurement methods, TRPM Cation Channels physiology
- Abstract
Menthol, the cooling natural product of peppermint, is widely used in medicinal preparations for the relief of acute and inflammatory pain in sports injuries, arthritis, and other painful conditions. Menthol induces the sensation of cooling by activating TRPM8, an ion channel in cold-sensitive peripheral sensory neurons. Recent studies identified additional targets of menthol, including the irritant receptor, TRPA1, voltage-gated ion channels and neurotransmitter receptors. It remains unclear which of these targets contribute to menthol-induced analgesia, or to the irritating side effects associated with menthol therapy. Here, we use genetic and pharmacological approaches in mice to probe the role of TRPM8 in analgesia induced by L-menthol, the predominant analgesic menthol isomer in medicinal preparations. L-menthol effectively diminished pain behavior elicited by chemical stimuli (capsaicin, acrolein, acetic acid), noxious heat, and inflammation (complete Freund's adjuvant). Genetic deletion of TRPM8 completely abolished analgesia by L-menthol in all these models, although other analgesics (acetaminophen) remained effective. Loss of L-menthol-induced analgesia was recapitulated in mice treated with a selective TRPM8 inhibitor, AMG2850. Selective activation of TRPM8 with WS-12, a menthol derivative that we characterized as a specific TRPM8 agonist in cultured sensory neurons and in vivo, also induced TRPM8-dependent analgesia of acute and inflammatory pain. L-menthol- and WS-12-induced analgesia was blocked by naloxone, suggesting activation of endogenous opioid-dependent analgesic pathways. Our data show that TRPM8 is the principal mediator of menthol-induced analgesia of acute and inflammatory pain. In contrast to menthol, selective TRPM8 agonists may produce analgesia more effectively, with diminished side effects., (Copyright © 2013 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
143. Sphingosine and FTY720 modulate pacemaking activity in interstitial cells of Cajal from mouse small intestine.
- Author
-
Nam JH, Kim WK, and Kim BJ
- Subjects
- Animals, Biological Clocks, Female, Fingolimod Hydrochloride, HEK293 Cells, Humans, Interstitial Cells of Cajal drug effects, Male, Membrane Potentials, Mice, Mice, Inbred BALB C, Organophosphates pharmacology, Patch-Clamp Techniques, Propylene Glycols pharmacology, TRPM Cation Channels drug effects, TRPM Cation Channels genetics, Chloride Channels physiology, Gastrointestinal Motility physiology, Interstitial Cells of Cajal physiology, Intestine, Small physiology, Sphingosine analogs & derivatives, Sphingosine pharmacology, TRPM Cation Channels physiology
- Abstract
Interstitial cells of Cajal (ICCs) are the pacemakers of the gastrointestinal tract, and transient receptor potential melastatin type 7 (TRPM7) and Ca(2+) activated Cl(-) channels (ANO1) are candidate the generators of pacemaker potentials in ICCs. The effects of D-erythro-sphingosine (SPH) and structural analogues of SPH, that is, N,N-dimethyl-Derythro-sphingosine (N,N-DMS), FTY720, and FTY720-P on the pacemaking activities of ICCs were examined using the whole cell patch clamp technique. SPH, N,N-DMS, and FTY720 decreased the amplitudes of pacemaker potentials in ICC clusters, but resting membrane potentials displayed little change. Also, perfusing SPH, N,N-DMS, or FTY720 in the bath reduced both inward and outward TRPM7-like currents in single ICCs, and inhibited ANO1 currents. The another structural analogue of SPH, FTY720-P was ineffective at the pacemaker potentials in ICC clusters and the TRPM7-like currents in single ICCs. Furthermore, FTY720-P had no effect on ANO1. These results suggest that SPH, N,N-DMS, and FTY720 modulate the pacemaker activities of ICCs, and that TRPM7 and ANO1 channels affect intestinal motility.
- Published
- 2013
- Full Text
- View/download PDF
144. TRPM7 is regulated by halides through its kinase domain.
- Author
-
Yu H, Zhang Z, Lis A, Penner R, and Fleig A
- Subjects
- Bromides metabolism, Cell Proliferation drug effects, Chlorides metabolism, DNA, Complementary biosynthesis, Dose-Response Relationship, Drug, HEK293 Cells, Humans, Hydrogen-Ion Concentration, Inhibitory Concentration 50, Iodides metabolism, MCF-7 Cells, Patch-Clamp Techniques, Protein Serine-Threonine Kinases, Protein Structure, Tertiary physiology, Real-Time Polymerase Chain Reaction, Symporters metabolism, TRPM Cation Channels physiology, Adenosine Triphosphate metabolism, Bromides pharmacology, Chlorides pharmacology, Feedback, Physiological physiology, Gene Expression Regulation physiology, Iodides pharmacology, TRPM Cation Channels metabolism
- Abstract
Transient receptor potential melastatin 7 (TRPM7) is a divalent-selective cation channel fused to an atypical α-kinase. TRPM7 is a key regulator of cell growth and proliferation, processes accompanied by mandatory cell volume changes. Osmolarity-induced cell volume alterations regulate TRPM7 through molecular crowding of solutes that affect channel activity, including magnesium (Mg(2+)), Mg-nucleotides and a further unidentified factor. Here, we assess whether chloride and related halides can act as negative feedback regulators of TRPM7. We find that chloride and bromide inhibit heterologously expressed TRPM7 in synergy with intracellular Mg(2+) ([Mg(2+)]i) and this is facilitated through the ATP-binding site of the channel's kinase domain. The synergistic block of TRPM7 by chloride and Mg(2+) is not reversed during divalent-free or acidic conditions, indicating a change in protein conformation that leads to channel inactivation. Iodide has the strongest inhibitory effect on TRPM7 at physiological [Mg(2+)]i. Iodide also inhibits endogenous TRPM7-like currents as assessed in MCF-7 breast cancer cells, where upregulation of SLC5A5 sodium-iodide symporter enhances iodide uptake and inhibits cell proliferation. These results indicate that chloride could be an important factor in modulating TRPM7 during osmotic stress and implicate TRPM7 as a possible molecular mechanism contributing to the anti-proliferative characteristics of intracellular iodide accumulation in cancer cells.
- Published
- 2013
- Full Text
- View/download PDF
145. Biological sex and mechanisms of ischemic brain injury.
- Author
-
Herson PS, Palmateer J, and Hurn PD
- Subjects
- Animals, Arachidonic Acids physiology, Cell Death physiology, Cells, Cultured, Disease Models, Animal, Epoxide Hydrolases, Female, Humans, Male, Nitric Oxide Synthase Type III physiology, Poly(ADP-ribose) Polymerases physiology, Signal Transduction, TRPM Cation Channels physiology, Brain Ischemia etiology, Sex Characteristics, Stroke etiology
- Abstract
Cerebrovascular disease is a leading cause of death-from-disease and of disability worldwide, affecting some 15 million people. The incidence of stroke or "brain attack" is unlikely to recede for a decade at minimum by most predictions, despite large public health initiatives in stroke prevention. It has been well established that stroke is also one of the most strikingly sex-specific diseases in its epidemiology, and in some cases, in patient outcomes. For example, women sustain lower rates of ischemic stroke relative to men, even beyond their menopausal years. In contrast, outcomes are worse in women in many clinical studies. The biological basis for this sexual dimorphism is a compelling story, and both hormone-dependent and hormone-independent factors are involved, the latter of which is the subject of this brief review. Understanding the molecular and cell-based mechanisms underlying sex differences in ischemic brain injury is an important step toward personalized medicine and effective therapeutic interventions in patients of both sexes.
- Published
- 2013
- Full Text
- View/download PDF
146. Oxidative stress-modulated TRPM ion channels in cell dysfunction and pathological conditions in humans.
- Author
-
Simon F, Varela D, and Cabello-Verrugio C
- Subjects
- Animals, Cardiovascular Diseases immunology, Cardiovascular Diseases metabolism, Cardiovascular Diseases pathology, Humans, Immunity, Innate, Neoplasms immunology, Neoplasms metabolism, Neoplasms pathology, Neurodegenerative Diseases immunology, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Phylogeny, Protein Conformation, TRPM Cation Channels chemistry, Oxidative Stress, TRPM Cation Channels physiology
- Abstract
The transient receptor potential melastatin (TRPM) protein family is an extensive group of ion channels expressed in several types of mammalian cells. Many studies have shown that these channels are crucial for performing several physiological functions. Additionally, a large body of evidence indicates that these channels are also involved in numerous human diseases, known as channelopathies. A characteristic event frequently observed during pathological states is the raising in intracellular oxidative agents over reducing molecules, shifting the redox balance and inducing oxidative stress. In particular, three members of the TRPM subfamily, TRPM2, TRPM4 and TRPM7, share the remarkable feature that their activities are modulated by oxidative stress. Because of the increase in oxidative stress, these TRPM channels function aberrantly, promoting the onset and development of diseases. Increases, absences, or modifications in the function of these redox-modulated TRPM channels are associated with cell dysfunction and human pathologies. Therefore, the effect of oxidative stress on ion channels becomes an essential part of the pathogenic mechanism. Thus, oxidative stress-modulated ion channels are more susceptible to generating pathological states than oxidant-independent channels. This review examines the most relevant findings regarding the participation of the oxidative stress-modulated TRPM ion channels, TRPM2, TRPM4, and TRPM7, in human diseases. In addition, the potential roles of these channels as therapeutic tools and targets for drug design are discussed., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
147. TRPM7 mediates breast cancer cell migration and invasion through the MAPK pathway.
- Author
-
Meng X, Cai C, Wu J, Cai S, Ye C, Chen H, Yang Z, Zeng H, Shen Q, and Zou F
- Subjects
- Cell Line, Tumor, Cell Movement, Female, Humans, Neoplasm Invasiveness, Phosphatidylinositol 3-Kinases physiology, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins c-akt physiology, RNA, Small Interfering genetics, Breast Neoplasms pathology, MAP Kinase Signaling System physiology, TRPM Cation Channels physiology
- Abstract
Metastasis is an inherent feature of breast cancer and transient receptor potential (TRP) channels were found to be potentially implicated in this process. Particularly, TRPM7 may regulate cell motility. We therefore examined the expression of TRPM7 mRNA in the Oncomine database and found that TRPM7 is correlated to metastasis and invasive breast cancer. Silencing TRPM7 with RNA interference resulted in a significant decrease in migration and invasion capability of MDA-MB-435 breast cancer cells, and phosphorylation levels of Src and MAPK but not AKT. Our results suggest that TRPM7 regulates migration and invasion of metastatic breast cancer cells via MAPK pathway., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
148. TRPM5-mediated calcium uptake regulates mucin secretion from human colon goblet cells.
- Author
-
Mitrovic S, Nogueira C, Cantero-Recasens G, Kiefer K, Fernández-Fernández JM, Popoff JF, Casano L, Bard FA, Gomez R, Valverde MA, and Malhotra V
- Subjects
- Colon cytology, Goblet Cells cytology, Golgi Apparatus drug effects, Golgi Apparatus metabolism, Humans, TRPM Cation Channels metabolism, Tetradecanoylphorbol Acetate pharmacology, Calcium metabolism, Colon metabolism, Goblet Cells metabolism, Mucins metabolism, TRPM Cation Channels physiology
- Abstract
Mucin 5AC (MUC5AC) is secreted by goblet cells of the respiratory tract and, surprisingly, also expressed de novo in mucus secreting cancer lines. siRNA-mediated knockdown of 7343 human gene products in a human colonic cancer goblet cell line (HT29-18N2) revealed new proteins, including a Ca(2+)-activated channel TRPM5, for MUC5AC secretion. TRPM5 was required for PMA and ATP-induced secretion of MUC5AC from the post-Golgi secretory granules. Stable knockdown of TRPM5 reduced a TRPM5-like current and ATP-mediated Ca(2+) signal. ATP-induced MUC5AC secretion depended strongly on Ca(2+) influx, which was markedly reduced in TRPM5 knockdown cells. The difference in ATP-induced Ca(2+) entry between control and TRPM5 knockdown cells was abrogated in the absence of extracellular Ca(2+) and by inhibition of the Na(+)/Ca(2+) exchanger (NCX). Accordingly, MUC5AC secretion was reduced by inhibition of NCX. Thus TRPM5 activation by ATP couples TRPM5-mediated Na(+) entry to promote Ca(2+) uptake via an NCX to trigger MUC5AC secretion. DOI:http://dx.doi.org/10.7554/eLife.00658.001.
- Published
- 2013
- Full Text
- View/download PDF
149. The relationship between α1-adrenergic receptors and TRPM8 channels in detrusor overactivity induced by cold stress in ovariectomized rats.
- Author
-
Noguchi W, Ishizuka O, Imamura T, Kurizaki Y, Yamagishi T, Yokoyama H, Lei Z, Silwal SG, Nishizawa O, and Andersson KE
- Subjects
- Animals, Cold Temperature, Female, Ovariectomy, Rats, Rats, Sprague-Dawley, Receptors, Adrenergic, alpha-1 physiology, Stress, Physiological, TRPM Cation Channels physiology, Urinary Bladder, Overactive etiology
- Abstract
Purpose: We studied whether cold stress induced detrusor overactivity in ovariectomized rats is associated with increased thermosensitive TRPM8 channel expression in the skin and whether the response could be inhibited by α1-adrenergic receptor blockade., Materials and Methods: A total of 24 Sprague-Dawley® rats at postnatal week 30 were randomly selected for ovariectomy (16) or sham ovariectomy (8). Five weeks later cystometric measurements of conscious, freely moving rats were made at room temperature (mean ± SEM 28C ± 2C) for 20 minutes. Eight ovariectomized rats were intravenously administered 1.0 mg/kg naftopidil. The other 8 ovariectomized and 8 sham operated rats were given naftopidil-free vehicle. Five minutes later they were transferred to a low temperature environment (mean 4C ± 2C) and micturition patterns were again recorded. TRPM8 channel expression in lumbar skin was estimated by real-time reverse-transcriptase polymerase chain reaction and immunohistochemistry., Results: TRPM8 channel mRNA and protein in the skin of ovariectomized rats were significantly higher than in sham operated rats. At room temperature micturition parameters were similar in sham operated and ovariectomized rats. At low temperature sham operated and ovariectomized rats showed cold stress induced detrusor overactivity but increased micturition frequency and decreased bladder capacity were significantly greater in ovariectomized rats. Treatment of ovariectomized rats with naftopidil inhibited cold stress induced detrusor overactivity., Conclusions: Cold stress induced detrusor overactivity in rats with decreased estrogen is associated with TRPM8 channel up-regulation in the skin and mediated by nerve pathways using α1-adrenergic receptors., (Copyright © 2013 American Urological Association Education and Research, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
150. Biochemical, single-channel, whole-cell patch clamp, and pharmacological analyses of endogenous TRPM4 channels in HEK293 cells.
- Author
-
Amarouch MY, Syam N, and Abriel H
- Subjects
- Calcium metabolism, Flufenamic Acid pharmacology, Humans, Patch-Clamp Techniques, Phenanthrenes pharmacology, TRPM Cation Channels antagonists & inhibitors, TRPM Cation Channels genetics, Transfection, TRPM Cation Channels physiology
- Abstract
Human embryonic kidney cells 293 (HEK293) are widely used as cellular heterologous expression systems to study transfected ion channels. This work characterizes the endogenous expression of TRPM4 channels in HEK293 cells. TRPM4 is an intracellular Ca(2+)-activated non-selective cationic channel expressed in many cell types. Western blot analyses have revealed the endogenous expression of TRPM4. Single channel 22pS conductance with a linear current-voltage relationship was observed using the inside-out patch clamp configuration in the presence of intracellular Ca(2+). The channels were permeable to the monovalent cations Na(+) and K(+), but not to Ca(2+). The open probability was voltage-dependent, being higher at positive potentials. Using the whole-cell patch clamp "ruptured patch" configuration, the amplitude of the intracellular Ca(2+)-activated macroscopic current was dependent on time after patch rupture. Initial transient activation followed by a steady-increase reaching a plateau phase was observed. Biophysical analyses of the macroscopic current showed common properties with those from HEK293 cells stably transfected with human TRPM4b, with the exception of current time course and Ca(2+) sensitivity. The endogenous macroscopic current reached the plateau faster and required 61.9±3.5μM Ca(2+) to be half-maximally activated versus 84.2±1.5μM for the transfected current. The pharmacological properties, however, were similar in both conditions. One hundred μM of flufenamic acid and 9-phenanthrol strongly inhibited the endogenous current. Altogether, the data demonstrate the expression of endogenous TRMP4 channels in HEK293 cells. This observation should be taken into account when using this cell line to study TRPM4 or other types of Ca(2+)-activated channels., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.