51
|
Steroids and TRP Channels: A Close Relationship. Int J Mol Sci 2020; 21:ijms21113819. [PMID: 32471309 PMCID: PMC7325571 DOI: 10.3390/ijms21113819] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) channels are remarkable transmembrane protein complexes that are essential for the physiology of the tissues in which they are expressed. They function as non-selective cation channels allowing for the signal transduction of several chemical, physical and thermal stimuli and modifying cell function. These channels play pivotal roles in the nervous and reproductive systems, kidney, pancreas, lung, bone, intestine, among others. TRP channels are finely modulated by different mechanisms: regulation of their function and/or by control of their expression or cellular/subcellular localization. These mechanisms are subject to being affected by several endogenously-produced compounds, some of which are of a lipidic nature such as steroids. Fascinatingly, steroids and TRP channels closely interplay to modulate several physiological events. Certain TRP channels are affected by the typical genomic long-term effects of steroids but others are also targets for non-genomic actions of some steroids that act as direct ligands of these receptors, as will be reviewed here.
Collapse
|
52
|
Van Hoeymissen E, Held K, Nogueira Freitas AC, Janssens A, Voets T, Vriens J. Gain of channel function and modified gating properties in TRPM3 mutants causing intellectual disability and epilepsy. eLife 2020; 9:57190. [PMID: 32427099 PMCID: PMC7253177 DOI: 10.7554/elife.57190] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEE) are a heterogeneous group of disorders characterized by epilepsy with comorbid intellectual disability. Recently, two de novo heterozygous mutations in the gene encoding TRPM3, a calcium permeable ion channel, were identified as the cause of DEE in eight probands, but the functional consequences of the mutations remained elusive. Here we demonstrate that both mutations (V990M and P1090Q) have distinct effects on TRPM3 gating, including increased basal activity, higher sensitivity to stimulation by the endogenous neurosteroid pregnenolone sulfate (PS) and heat, and altered response to ligand modulation. Most strikingly, the V990M mutation affected the gating of the non-canonical pore of TRPM3, resulting in large inward cation currents via the voltage sensor domain in response to PS stimulation. Taken together, these data indicate that the two DEE mutations in TRPM3 result in a profound gain of channel function, which may lie at the basis of epileptic activity and neurodevelopmental symptoms in the patients.
Collapse
Affiliation(s)
- Evelien Van Hoeymissen
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, Leuven, Belgium.,Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Belgium and Department of Molecular Medicine, Leuven, Belgium
| | - Katharina Held
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, Leuven, Belgium.,Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Belgium and Department of Molecular Medicine, Leuven, Belgium
| | - Ana Cristina Nogueira Freitas
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Belgium and Department of Molecular Medicine, Leuven, Belgium
| | - Annelies Janssens
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Belgium and Department of Molecular Medicine, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, Belgium and Department of Molecular Medicine, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, Leuven, Belgium
| |
Collapse
|
53
|
Zhao S, Yudin Y, Rohacs T. Disease-associated mutations in the human TRPM3 render the channel overactive via two distinct mechanisms. eLife 2020; 9:e55634. [PMID: 32343227 PMCID: PMC7255801 DOI: 10.7554/elife.55634] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022] Open
Abstract
Transient Receptor Potential Melastatin 3 (TRPM3) is a Ca2+ permeable non-selective cation channel activated by heat and chemical agonists such as pregnenolone sulfate and CIM0216. TRPM3 mutations in humans were recently reported to be associated with intellectual disability and epilepsy; the functional effects of those mutations, however, were not reported. Here, we show that both disease-associated mutations in the human TRPM3 render the channel overactive, but likely via different mechanisms. The Val to Met substitution in the S4-S5 loop induced a larger increase in basal activity and agonist sensitivity at room temperature than the Pro to Gln substitution in the extracellular segment of S6. In contrast, heat activation was increased more by the S6 mutant than by the S4-S5 segment mutant. Both mutants were inhibited by the TRPM3 antagonist primidone, suggesting a potential therapeutic intervention to treat this disease.
Collapse
Affiliation(s)
- Siyuan Zhao
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| | - Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| |
Collapse
|
54
|
Webster CM, Tworig J, Caval-Holme F, Morgans CW, Feller MB. The Impact of Steroid Activation of TRPM3 on Spontaneous Activity in the Developing Retina. eNeuro 2020; 7:ENEURO.0175-19.2020. [PMID: 32238415 PMCID: PMC7177749 DOI: 10.1523/eneuro.0175-19.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 02/19/2020] [Accepted: 03/06/2020] [Indexed: 12/19/2022] Open
Abstract
In the central nervous system, melastatin transient receptor potential (TRPM) channels function as receptors for the neurosteroid pregnenolone sulfate (PregS). The expression and function of TRPM3 has been explored in adult retina, although its role during development is unknown. We found, during the second postnatal week in mice, TRPM3 immunofluorescence labeled distinct subsets of inner retinal neurons, including a subset of retinal ganglion cells (RGCs), similar to what has been reported in the adult. Labeling for a TRPM3 promoter-driven reporter confirmed expression of the TRPM3 gene in RGCs and revealed additional expression in nearly all Müller glial cells. Using two-photon calcium imaging, we show that PregS and the synthetic TRPM3 agonist CIM0216 (CIM) induced prolonged calcium transients in RGCs, which were mostly absent in TRPM3 knock-out (KO) mice. These prolonged calcium transients were not associated with strong membrane depolarizations but induced c-Fos expression. To elucidate the impact of PregS-activation of TRPM3 on retinal circuits we took two sets of physiological measurements. First, PregS induced a robust increase in the frequency but not amplitude of spontaneous postsynaptic currents (PSCs). This increase was absent in the TRPM3 KO mice. Second, PregS induced a small increase in cell participation and duration of retinal waves, but this modulation persisted in TRPM3 KO mice, indicating PregS was acting on wave generating circuits independent of TRPM3 channels. Though baseline frequency of retinal waves was slightly reduced in the TRPM3 KO mice, other properties of waves were indistinguishable from wildtype. Together, these results indicate that the presence of neurosteroids impact spontaneous synaptic activity and retinal waves during development via both TRPM3-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Corey M Webster
- Department of Molecular and Cell Biology, University of California. Berkeley, Berkeley, CA 94720-3200
| | - Joshua Tworig
- Department of Molecular and Cell Biology, University of California. Berkeley, Berkeley, CA 94720-3200
| | - Franklin Caval-Holme
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3200
| | - Catherine W Morgans
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California. Berkeley, Berkeley, CA 94720-3200
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3200
| |
Collapse
|
55
|
Islam MS. Molecular Regulations and Functions of the Transient Receptor Potential Channels of the Islets of Langerhans and Insulinoma Cells. Cells 2020; 9:cells9030685. [PMID: 32168890 PMCID: PMC7140661 DOI: 10.3390/cells9030685] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 12/17/2022] Open
Abstract
Insulin secretion from the β-cells of the islets of Langerhans is triggered mainly by nutrients such as glucose, and incretin hormones such as glucagon-like peptide-1 (GLP-1). The mechanisms of the stimulus-secretion coupling involve the participation of the key enzymes that metabolize the nutrients, and numerous ion channels that mediate the electrical activity. Several members of the transient receptor potential (TRP) channels participate in the processes that mediate the electrical activities and Ca2+ oscillations in these cells. Human β-cells express TRPC1, TRPM2, TRPM3, TRPM4, TRPM7, TRPP1, TRPML1, and TRPML3 channels. Some of these channels have been reported to mediate background depolarizing currents, store-operated Ca2+ entry (SOCE), electrical activity, Ca2+ oscillations, gene transcription, cell-death, and insulin secretion in response to stimulation by glucose and GLP1. Different channels of the TRP family are regulated by one or more of the following mechanisms: activation of G protein-coupled receptors, the filling state of the endoplasmic reticulum Ca2+ store, heat, oxidative stress, or some second messengers. This review briefly compiles our current knowledge about the molecular mechanisms of regulations, and functions of the TRP channels in the β-cells, the α-cells, and some insulinoma cell lines.
Collapse
Affiliation(s)
- Md. Shahidul Islam
- Karolinska Institutet, Department of Clinical Science and Education, Södersjukhuset, Research Center, 5th floor, SE-118 83 Stockholm, Sweden;
- Department of Emergency Care and Internal Medicine, Uppsala University Hospital, Uppsala University, SE-751 85 Uppsala, Sweden
| |
Collapse
|
56
|
Vangeel L, Benoit M, Miron Y, Miller PE, De Clercq K, Chaltin P, Verfaillie C, Vriens J, Voets T. Functional expression and pharmacological modulation of TRPM3 in human sensory neurons. Br J Pharmacol 2020; 177:2683-2695. [PMID: 31985045 DOI: 10.1111/bph.14994] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/09/2019] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE The transient receptor potential (TRP) ion channel TRPM3 functions as a noxious heat sensor, plays a key role in acute pain sensation and inflammatory hyperalgesia in rodents. Despite its potential as a novel analgesic drug target, little is known about the expression, function and modulation in the humans. EXPERIMENTAL APPROACH We studied TRPM3 in freshly isolated human dorsal root ganglion (hDRG) neurons and human stem cell-derived sensory (hSCDS) neurons. Expression was analysed at the mRNA level using RT-qPCR. Channel function was assessed using Fura-2-based calcium imaging and whole-cell patch-clamp recordings. KEY RESULTS TRPM3 was detected at the mRNA level in both hDRG and hSCDS neurons. The TRPM3 agonists pregnenolone sulphate (PS) and CIM0216 evoked robust intracellular Ca2+ responses in 52% of hDRG and 58% of hSCDS neurons. Whole-cell patch-clamp recordings in hSCDS neurons revealed pregnenolone sulphate (PS)- and CIM0216-evoked currents exhibiting the characteristic current-voltage relation of TRPM3. PS-induced calcium responses in hSCDS neurons were reversed in a dose-dependent manner by the flavonoid isosakuranetin and by antiseizure drug primidone. Finally, the μ-opioid receptor agonist DAMGO and the GABAB receptor agonist baclofen inhibited PS-evoked TRPM3 responses in a subset of hSCDS neurons. CONCLUSION AND IMPLICATIONS These results provide the first direct evidence of functional expression of the pain receptor TRPM3 in human sensory neurons, largely mirroring the channel's properties observed in mouse sensory neurons. hSCDS neurons represent a valuable and readily accessible in vitro model to study TRPM3 regulation and pharmacology in a relevant human cellular context.
Collapse
Affiliation(s)
- Laura Vangeel
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Melissa Benoit
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | | | | - Katrien De Clercq
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, G-PURE, KU Leuven, Leuven, Belgium
| | - Patrick Chaltin
- Center for Drug Design and Discovery, Bio-Incubator 2, Heverlee, Belgium
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, G-PURE, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
57
|
Shiels A. TRPM3_miR-204: a complex locus for eye development and disease. Hum Genomics 2020; 14:7. [PMID: 32070426 PMCID: PMC7027284 DOI: 10.1186/s40246-020-00258-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
First discovered in a light-sensitive retinal mutant of Drosophila, the transient receptor potential (TRP) superfamily of non-selective cation channels serve as polymodal cellular sensors that participate in diverse physiological processes across the animal kingdom including the perception of light, temperature, pressure, and pain. TRPM3 belongs to the melastatin sub-family of TRP channels and has been shown to function as a spontaneous calcium channel, with permeability to other cations influenced by alternative splicing and/or non-canonical channel activity. Activators of TRPM3 channels include the neurosteroid pregnenolone sulfate, calmodulin, phosphoinositides, and heat, whereas inhibitors include certain drugs, plant-derived metabolites, and G-protein subunits. Activation of TRPM3 channels at the cell membrane elicits a signal transduction cascade of mitogen-activated kinases and stimulus response transcription factors. The mammalian TRPM3 gene hosts a non-coding microRNA gene specifying miR-204 that serves as both a tumor suppressor and a negative regulator of post-transcriptional gene expression during eye development in vertebrates. Ocular co-expression of TRPM3 and miR-204 is upregulated by the paired box 6 transcription factor (PAX6) and mutations in all three corresponding genes underlie inherited forms of eye disease in humans including early-onset cataract, retinal dystrophy, and coloboma. This review outlines the genomic and functional complexity of the TRPM3_miR-204 locus in mammalian eye development and disease.
Collapse
Affiliation(s)
- Alan Shiels
- Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Ave., Box 8096, St. Louis, MO, 63110, USA.
| |
Collapse
|
58
|
Behrendt M. Transient receptor potential channels in the context of nociception and pain - recent insights into TRPM3 properties and function. Biol Chem 2020; 400:917-926. [PMID: 30844758 DOI: 10.1515/hsz-2018-0455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/25/2019] [Indexed: 01/09/2023]
Abstract
Potential harmful stimuli like heat, mechanical pressure or chemicals are detected by specialized cutaneous nerve fiber endings of nociceptor neurons in a process called nociception. Acute stimulation results in immediate protective reflexes and pain sensation as a normal, physiological behavior. However, ongoing (chronic) pain is a severe pathophysiological condition with diverse pathogeneses that is clinically challenging because of limited therapeutic options. Therefore, an urgent need exists for new potent and specific analgesics without afflicting adverse effects. Recently, TRPM3, a member of the superfamily of transient receptor potential (TRP) ion channels, has been shown to be expressed in nociceptors and to be involved in the detection of noxious heat (acute pain) as well as inflammatory hyperalgesia (acute and chronic pain). Current results in TRPM3 research indicate that this ion channel might not only be part of yet unraveled mechanisms underlying chronic pain but also has the potential to become a clinically relevant pharmacological target of future analgesic strategies. The aim of this review is to summarize and present the basic features of TRPM3 proteins and channels, to highlight recent findings and developments and to provide an outlook on emerging directions of TRPM3 research in the field of chronic pain.
Collapse
Affiliation(s)
- Marc Behrendt
- Experimental Pain Research, Heidelberg University, Medical Faculty Mannheim, CBTM, Tridomus, Building C, Ludolf-Krehl-Straße 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
59
|
Kelemen B, Lisztes E, Vladár A, Hanyicska M, Almássy J, Oláh A, Szöllősi AG, Pénzes Z, Posta J, Voets T, Bíró T, Tóth BI. Volatile anaesthetics inhibit the thermosensitive nociceptor ion channel transient receptor potential melastatin 3 (TRPM3). Biochem Pharmacol 2020; 174:113826. [PMID: 31987857 DOI: 10.1016/j.bcp.2020.113826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/22/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Volatile anaesthetics (VAs) are the most widely used compounds to induce reversible loss of consciousness and maintain general anaesthesia during surgical interventions. Although the mechanism of their action is not yet fully understood, it is generally believed, that VAs depress central nervous system functions mainly through modulation of ion channels in the neuronal membrane, including 2-pore-domain K+ channels, GABA and NMDA receptors. Recent research also reported their action on nociceptive and thermosensitive TRP channels expressed in the peripheral nervous system, including TRPV1, TRPA1, and TRPM8. Here, we investigated the effect of VAs on TRPM3, a less characterized member of the thermosensitive TRP channels playing a central role in noxious heat sensation. METHODS We investigated the effect of VAs on the activity of recombinant and native TRPM3, by monitoring changes in the intracellular Ca2+ concentration and measuring TRPM3-mediated transmembrane currents. RESULTS All the investigated VAs (chloroform, halothane, isoflurane, sevoflurane) inhibited both the agonist-induced (pregnenolone sulfate, CIM0216) and heat-activated Ca2+ signals and transmembrane currents in a concentration dependent way in HEK293T cells overexpressing recombinant TRPM3. Among the tested VAs, halothane was the most potent blocker (IC50 = 0.52 ± 0.05 mM). We also investigated the effect of VAs on native TRPM3 channels expressed in sensory neurons of the dorsal root ganglia. While VAs activated certain sensory neurons independently of TRPM3, they strongly and reversibly inhibited the agonist-induced TRPM3 activity. CONCLUSIONS These data provide a better insight into the molecular mechanism beyond the analgesic effect of VAs and propose novel strategies to attenuate TRPM3 dependent nociception.
Collapse
Affiliation(s)
- Balázs Kelemen
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Erika Lisztes
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Vladár
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Martin Hanyicska
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - János Almássy
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Gábor Szöllősi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pénzes
- Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, Hungary; Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Posta
- Laboratory of Toxicology, Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine and TRP Research Platform Leuven (TRPLe), KU Leuven, Leuven, Belgium
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Hungarian Center of Excellence for Molecular Medicine, Szeged, Hungary
| | - Balázs István Tóth
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
60
|
Alkhatib O, da Costa R, Gentry C, Quallo T, Bevan S, Andersson DA. Promiscuous G-Protein-Coupled Receptor Inhibition of Transient Receptor Potential Melastatin 3 Ion Channels by Gβγ Subunits. J Neurosci 2019; 39:7840-7852. [PMID: 31451581 PMCID: PMC6774412 DOI: 10.1523/jneurosci.0882-19.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/19/2019] [Accepted: 08/14/2019] [Indexed: 11/21/2022] Open
Abstract
Transient receptor potential melastatin 3 (TRPM3) is a nonselective cation channel that is inhibited by Gβγ subunits liberated following activation of Gαi/o protein-coupled receptors. Here, we demonstrate that TRPM3 channels are also inhibited by Gβγ released from Gαs and Gαq Activation of the Gs-coupled adenosine 2B receptor and the Gq-coupled muscarinic acetylcholine M1 receptor inhibited the activity of TRPM3 heterologously expressed in HEK293 cells. This inhibition was prevented when the Gβγ sink βARK1-ct (C terminus of β-adrenergic receptor kinase-1) was coexpressed with TRPM3. In neurons isolated from mouse dorsal root ganglion (DRG), native TRPM3 channels were inhibited by activating Gs-coupled prostaglandin-EP2 and Gq-coupled bradykinin B2 (BK2) receptors. The Gi/o inhibitor pertussis toxin and inhibitors of PKA and PKC had no effect on EP2- and BK2-mediated inhibition of TRPM3, demonstrating that the receptors did not act through Gαi/o or through the major protein kinases activated downstream of G-protein-coupled receptor (GPCR) activation. When DRG neurons were dialyzed with GRK2i, which sequesters free Gβγ protein, TRPM3 inhibition by EP2 and BK2 was significantly reduced. Intraplantar injections of EP2 or BK2 agonists inhibited both the nocifensive response evoked by TRPM3 agonists, and the heat hypersensitivity produced by Freund's Complete Adjuvant (FCA). Furthermore, FCA-induced heat hypersensitivity was completely reversed by the selective TRPM3 antagonist ononetin in WT mice and did not develop in Trpm3-/- mice. Our results demonstrate that TRPM3 is subject to promiscuous inhibition by Gβγ protein in heterologous expression systems, primary neurons and in vivo, and suggest a critical role for this ion channel in inflammatory heat hypersensitivity.SIGNIFICANCE STATEMENT The ion channel TRPM3 is widely expressed in the nervous system. Recent studies showed that Gαi/o-coupled GPCRs inhibit TRPM3 through a direct interaction between Gβγ subunits and TRPM3. Since Gβγ proteins can be liberated from other Gα subunits than Gαi/o, we examined whether activation of Gs- and Gq-coupled receptors also influence TRPM3 via Gβγ. Our results demonstrate that activation of Gs- and Gq-coupled GPCRs in recombinant cells and sensory neurons inhibits TRPM3 via Gβγ liberation. We also demonstrated that Gs- and Gq-coupled receptors inhibit TRPM3 in vivo, thereby reducing pain produced by activation of TRPM3, and inflammatory heat hypersensitivity. Our results identify Gβγ inhibition of TRPM3 as an effector mechanism shared by the major Gα subunits.
Collapse
Affiliation(s)
- Omar Alkhatib
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom, and
| | - Robson da Costa
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom, and
- School of Pharmacy, Universidade Federal do Rio de Janeiro, 21941-908 Rio de Janeiro, Brazil
| | - Clive Gentry
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom, and
| | - Talisia Quallo
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom, and
| | - Stuart Bevan
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom, and
| | - David A Andersson
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom, and
| |
Collapse
|
61
|
Wong KK, Banham AH, Yaacob NS, Nur Husna SM. The oncogenic roles of TRPM ion channels in cancer. J Cell Physiol 2019; 234:14556-14573. [PMID: 30710353 DOI: 10.1002/jcp.28168] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Transient receptor potential (TRP) proteins are a diverse family of ion channels present in multiple types of tissues. They function as gatekeepers for responses to sensory stimuli including temperature, vision, taste, and pain through their activities in conducting ion fluxes. The TRPM (melastatin) subfamily consists of eight members (i.e., TRPM1-8), which collectively regulate fluxes of various types of cations such as K+ , Na+ , Ca2+ , and Mg2+ . Growing evidence in the past two decades indicates that TRPM ion channels, their isoforms, or long noncoding RNAs encoded within the locus may be oncogenes involved in the regulation of cancer cell growth, proliferation, autophagy, invasion, and epithelial-mesenchymal transition, and their significant association with poor clinical outcomes of cancer patients. In this review, we describe and discuss recent findings implicating TRPM channels in different malignancies, their functions, mechanisms, and signaling pathways involved in cancers, as well as summarizing their normal physiological functions and the availability of ion channel pharmacological inhibitors.
Collapse
Affiliation(s)
- Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Alison H Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
62
|
Voets T, Vriens J, Vennekens R. Targeting TRP Channels - Valuable Alternatives to Combat Pain, Lower Urinary Tract Disorders, and Type 2 Diabetes? Trends Pharmacol Sci 2019; 40:669-683. [PMID: 31395287 DOI: 10.1016/j.tips.2019.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/12/2019] [Accepted: 07/10/2019] [Indexed: 12/18/2022]
Abstract
Transient receptor potential (TRP) channels are a family of functionally diverse and widely expressed cation channels involved in a variety of cell signaling and sensory pathways. Research in the last two decades has not only shed light on the physiological roles of the 28 mammalian TRP channels, but also revealed the involvement of specific TRP channels in a plethora of inherited and acquired human diseases. Considering the historical successes of other types of ion channels as therapeutic drug targets, small molecules that target specific TRP channels hold promise as treatments for a variety of human conditions. In recent research, important new findings have highlighted the central role of TRP channels in chronic pain, lower urinary tract disorders, and type 2 diabetes, conditions with an unmet medical need. Here, we discuss how these advances support the development of TRP-channel-based pharmacotherapies as valuable alternatives to the current mainstays of treatment.
Collapse
Affiliation(s)
- Thomas Voets
- Laboratory of Ion Channel Research, VIB Center for Brain and Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, VIB Center for Brain and Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
63
|
Takayama Y, Derouiche S, Maruyama K, Tominaga M. Emerging Perspectives on Pain Management by Modulation of TRP Channels and ANO1. Int J Mol Sci 2019; 20:E3411. [PMID: 31336748 PMCID: PMC6678529 DOI: 10.3390/ijms20143411] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022] Open
Abstract
Receptor-type ion channels are critical for detection of noxious stimuli in primary sensory neurons. Transient receptor potential (TRP) channels mediate pain sensations and promote a variety of neuronal signals that elicit secondary neural functions (such as calcitonin gene-related peptide [CGRP] secretion), which are important for physiological functions throughout the body. In this review, we focus on the involvement of TRP channels in sensing acute pain, inflammatory pain, headache, migraine, pain due to fungal infections, and osteo-inflammation. Furthermore, action potentials mediated via interactions between TRP channels and the chloride channel, anoctamin 1 (ANO1), can also generate strong pain sensations in primary sensory neurons. Thus, we also discuss mechanisms that enhance neuronal excitation and are dependent on ANO1, and consider modulation of pain sensation from the perspective of both cation and anion dynamics.
Collapse
Affiliation(s)
- Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Sandra Derouiche
- Thermal Biology group, Exploratory Research Center on Life and Living Systems, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| | - Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| | - Makoto Tominaga
- Thermal Biology group, Exploratory Research Center on Life and Living Systems, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
64
|
Yudin Y, Rohacs T. The G-protein-biased agents PZM21 and TRV130 are partial agonists of μ-opioid receptor-mediated signalling to ion channels. Br J Pharmacol 2019; 176:3110-3125. [PMID: 31074038 DOI: 10.1111/bph.14702] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/10/2019] [Accepted: 04/13/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Opioids remain the most efficient medications against severe pain; they act on receptors that couple to heterotrimeric G-proteins in the Gαi/o family. Opioids exert many of their acute effects through modulating ion channels via Gβγ subunits. Many of their side effects are attributed to β-arrestin recruitment. Several biased agonists that do not recruit β-arrestins, but activate G-protein-dependent pathways, have recently been developed. While these compounds have been proposed to be full agonists of G-protein signalling in several high throughput pharmacological assays, their effects were not studied on ion channel targets. EXPERIMENTAL APPROACH Here, we used patch-clamp electrophysiology and Ca2+ imaging to test the effects of TRV130, PZM21, and herkinorin, three G-protein-biased agonists of μ-opioid receptors, on ion channel targets of Gαi/o /Gβγ signalling. We also studied G-protein dissociation using a FRET-based assay. KEY RESULTS All three biased agonists induced smaller activation of G-protein-coupled inwardly rectifying K+ channels (Kir 3.2) and smaller inhibition of transient receptor potential melastatin (TRPM3) channels than the full μ receptor agonist DAMGO. Co-application of TRV130 or PZM21, but not herkinorin, alleviated the effects of DAMGO on both channels. PZM21 and TRV130 also decreased the effect of morphine on Kir 3.2 channels. The CaV 2.2 channel was also inhibited less by PZM21 and TRV130 than by DAMGO. We also found that TRV130, PZM21, and herkinorin were less effective than DAMGO at inducing dissociation of the Gαi /Gβγ complex. CONCLUSION AND IMPLICATIONS TRV130, PZM21, and potentially herkinorin are partial agonists of μ receptors.
Collapse
Affiliation(s)
- Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
65
|
Sarmiento BE, Santos Menezes LF, Schwartz EF. Insulin Release Mechanism Modulated by Toxins Isolated from Animal Venoms: From Basic Research to Drug Development Prospects. Molecules 2019; 24:E1846. [PMID: 31091684 PMCID: PMC6571724 DOI: 10.3390/molecules24101846] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/23/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Venom from mammals, amphibians, snakes, arachnids, sea anemones and insects provides diverse sources of peptides with different potential medical applications. Several of these peptides have already been converted into drugs and some are still in the clinical phase. Diabetes type 2 is one of the diseases with the highest mortality rate worldwide, requiring specific attention. Diverse drugs are available (e.g., Sulfonylureas) for effective treatment, but with several adverse secondary effects, most of them related to the low specificity of these compounds to the target. In this context, the search for specific and high-affinity compounds for the management of this metabolic disease is growing. Toxins isolated from animal venom have high specificity and affinity for different molecular targets, of which the most important are ion channels. This review will present an overview about the electrical activity of the ion channels present in pancreatic β cells that are involved in the insulin secretion process, in addition to the diversity of peptides that can interact and modulate the electrical activity of pancreatic β cells. The importance of prospecting bioactive peptides for therapeutic use is also reinforced.
Collapse
Affiliation(s)
- Beatriz Elena Sarmiento
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| | - Luis Felipe Santos Menezes
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| | - Elisabeth F Schwartz
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| |
Collapse
|
66
|
Alonso-Carbajo L, Alpizar YA, Startek JB, López-López JR, Pérez-García MT, Talavera K. Activation of the cation channel TRPM3 in perivascular nerves induces vasodilation of resistance arteries. J Mol Cell Cardiol 2019; 129:219-230. [PMID: 30853321 DOI: 10.1016/j.yjmcc.2019.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
The Transient Receptor Potential Melastatin 3 (TRPM3) is a Ca2+-permeable non-selective cation channel activated by the neurosteroid pregnenolone sulfate (PS). This compound was previously shown to contract mouse aorta by activating TRPM3 in vascular smooth muscle cells (VSMC), and proposed as therapeutic modulator of vascular functions. However, PS effects and the role of TRPM3 in resistance arteries remain unknown. Thus, we aimed at determining the localization and physiological role of TRPM3 in mouse mesenteric arteries. Real-time qPCR experiments, anatomical localization using immunofluorescence microscopy and patch-clamp recordings in isolated VSMC showed that TRPM3 expression in mesenteric arteries is restricted to perivascular nerves. Pressure myography experiments in wild type (WT) mouse arteries showed that PS vasodilates with a concentration-dependence that was best fit by two Hill components (effective concentrations, EC50, of 14 and 100 μM). The low EC50 component was absent in preparations from Trpm3 knockout (KO) mice and in WT arteries in the presence of the CGRP receptor antagonist BIBN 4096. TRPM3-dependent vasodilation was partially inhibited by a cocktail of K+ channel blockers, and not mediated by β-adrenergic signaling. We conclude that, contrary to what was found in aorta, PS dilates mesenteric arteries, partly via an activation of TRPM3 that triggers CGRP release from perivascular nerve endings and a subsequent activation of K+ channels in VSMC. We propose that TRPM3 is implicated in the regulation of the tone of resistance arteries and that its activation by yet unidentified endogenous damage-associated molecules lead to protective vasodilation responses in mesenteric arteries.
Collapse
Affiliation(s)
- Lucía Alonso-Carbajo
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium; Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Universidad de Valladolid y CSIC, Sanz y Forés 3, 47003 Valladolid, Spain
| | - Yeranddy A Alpizar
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium
| | - Justyna B Startek
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium
| | - José Ramón López-López
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Universidad de Valladolid y CSIC, Sanz y Forés 3, 47003 Valladolid, Spain
| | - María Teresa Pérez-García
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Universidad de Valladolid y CSIC, Sanz y Forés 3, 47003 Valladolid, Spain
| | - Karel Talavera
- Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49, Campus Gasthuisberg, O&N1 Box 802, 3000 Leuven, Belgium.
| |
Collapse
|
67
|
Transient receptor potential melastatin-3 in the rat sensory ganglia of the trigeminal, glossopharyngeal and vagus nerves. J Chem Neuroanat 2019; 96:116-125. [PMID: 30639448 DOI: 10.1016/j.jchemneu.2019.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Transient receptor potential melastatin-3 (TRPM3) is a nonselective cation channel, has permeability of Ca2+, and probably participates in thermosensitive nociception. In this study, immunohistochemistry for TRPM3 was conducted in the rat trigeminal, glossopharyngeal and vagal sensory ganglia. TRPM3-immunoreactivity was expressed by half of sensory neurons in the trigeminal (TG), petrosal (PG) and jugular ganglia (JG), and by about 80% of sensory neurons in the nodose ganglion (NG). They mostly had small to medium-sized cell bodies. A trichrome immunofluorescence method showed co-existence of TRPM3 with TRP vanilloid 1 (TRPV1) and calcitonin gene-related peptide (CGRP). Approximately 70% of TRPM3-immunoreactive (-IR) neurons contained TRPV1-immunoreactivity in all the examined ganglia. More than 40% of TRPM3-IR neurons exhibited CGRP-immunoreactivity in the TG, PG and JG. Only a few sensory neurons co-expressed TRPM3- and CGRP-immunoreactivity in the NG. In addition, more than 40% of TRPM3-IR neurons bound to isolectin B4 in all the examined ganglia. By combination of retrograde tracing method and immunohistochemistry, half of TG neurons innervating the facial skin and incisive papilla expressed TRPM3-immunoreactivity whereas approximately 20% of those innervating the tooth pulp contained TRPM3-immunoreactivity. Co-expression of TRPM3-immunoreactivity with TRPV1- or CGRP-immunoreactivity was common among cutaneous and papillary TG neurons but not among pulpal TG neurons. More than 60% of PG and JG neurons innervating the external ear canal skin and circumvallate papilla contained TRPM3-immunoreactivity. Co-expression of TRPM3 with TRPV1 or CGRP was common among PG and JG neurons innervating the external ear canal skin. However, a smaller number of TRPM3-IR neurons co-expressing TRPV1- or CGRP-immunoreactivity innervate the circumvallate papilla in the PG. The present study suggests that expression of TRPM3 and its co-existence with TRPV1 and CGRP in sensory neurons depend on the variety of their peripheral targets in the trigeminal, glossopharyngeal and vagal nervous systems.
Collapse
|
68
|
Yudin Y, Rohacs T. Inhibitory G i/O-coupled receptors in somatosensory neurons: Potential therapeutic targets for novel analgesics. Mol Pain 2018; 14:1744806918763646. [PMID: 29580154 PMCID: PMC5882016 DOI: 10.1177/1744806918763646] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Primary sensory neurons in the dorsal root ganglia and trigeminal ganglia are responsible for sensing mechanical and thermal stimuli, as well as detecting tissue damage. These neurons express ion channels that respond to thermal, mechanical, or chemical cues, conduct action potentials, and mediate transmitter release. These neurons also express a large number of G-protein coupled receptors, which are major transducers for extracellular signaling molecules, and their activation usually modulates the primary transduction pathways. Receptors that couple to phospholipase C via heterotrimeric Gq/11 proteins and those that activate adenylate cyclase via Gs are considered excitatory; they positively regulate somatosensory transduction and they play roles in inflammatory sensitization and pain, and in some cases also in inducing itch. On the other hand, receptors that couple to Gi/o proteins, such as opioid or GABAB receptors, are generally inhibitory. Their activation counteracts the effect of Gs-stimulation by inhibiting adenylate cyclase, as well as exerts effects on ion channels, usually resulting in decreased excitability. This review will summarize knowledge on Gi-coupled receptors in sensory neurons, focusing on their roles in ion channel regulation and discuss their potential as targets for analgesic and antipruritic medications.
Collapse
Affiliation(s)
- Yevgen Yudin
- 1 Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Tibor Rohacs
- 1 Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
69
|
Bishnoi M, Khare P, Brown L, Panchal SK. Transient receptor potential (TRP) channels: a metabolic TR(i)P to obesity prevention and therapy. Obes Rev 2018; 19:1269-1292. [PMID: 29797770 DOI: 10.1111/obr.12703] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/26/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022]
Abstract
Cellular transport of ions, especially by ion channels, regulates physiological function. The transient receptor potential (TRP) channels, with 30 identified so far, are cation channels with high calcium permeability. These ion channels are present in metabolically active tissues including adipose tissue, liver, gastrointestinal tract, brain (hypothalamus), pancreas and skeletal muscle, which suggests a potential role in metabolic disorders including obesity. TRP channels have potentially important roles in adipogenesis, obesity development and its prevention and therapy because of their physiological properties including calcium permeability, thermosensation and taste perception, involvement in cell metabolic signalling and hormone release. This wide range of actions means that organ-specific actions are unlikely, thus increasing the possibility of adverse effects. Delineation of responses to TRP channels has been limited by the poor selectivity of available agonists and antagonists. Food constituents that can modulate TRP channels are of interest in controlling metabolic status. TRP vanilloid 1 channels modulated by capsaicin have been the most studied, suggesting that this may be the first target for effective pharmacological modulation in obesity. This review shows that most of the TRP channels are potential targets to reduce metabolic disorders through a range of mechanisms.
Collapse
Affiliation(s)
- M Bishnoi
- Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, S.A.S. Nagar (Mohali), Punjab, India.,Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia
| | - P Khare
- Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, S.A.S. Nagar (Mohali), Punjab, India
| | - L Brown
- Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia.,School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | - S K Panchal
- Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia
| |
Collapse
|
70
|
Boonen B, Alpizar YA, Meseguer VM, Talavera K. TRP Channels as Sensors of Bacterial Endotoxins. Toxins (Basel) 2018; 10:toxins10080326. [PMID: 30103489 PMCID: PMC6115757 DOI: 10.3390/toxins10080326] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 02/07/2023] Open
Abstract
The cellular and systemic effects induced by bacterial lipopolysaccharides (LPS) have been solely attributed to the activation of the Toll-like receptor 4 (TLR4) signalling cascade. However, recent studies have shown that LPS activates several members of the Transient Receptor Potential (TRP) family of cation channels. Indeed, LPS induces activation of the broadly-tuned chemosensor TRPA1 in sensory neurons in a TLR4-independent manner, and genetic ablation of this channel reduced mouse pain and inflammatory responses triggered by LPS and the gustatory-mediated avoidance to LPS in fruit flies. LPS was also shown to activate TRPV4 channels in airway epithelial cells, an effect leading to an immediate production of bactericidal nitric oxide and to an increase in ciliary beat frequency. In this review, we discuss the role of TRP channels as sensors of bacterial endotoxins, and therefore, as crucial players in the timely detection of invading gram-negative bacteria.
Collapse
Affiliation(s)
- Brett Boonen
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, O&N1 Herestraat 49 - box 802, 3000 Leuven, Belgium.
| | - Yeranddy A Alpizar
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, O&N1 Herestraat 49 - box 802, 3000 Leuven, Belgium.
| | - Victor M Meseguer
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain.
| | - Karel Talavera
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, O&N1 Herestraat 49 - box 802, 3000 Leuven, Belgium.
| |
Collapse
|
71
|
Held K, Gruss F, Aloi VD, Janssens A, Ulens C, Voets T, Vriens J. Mutations in the voltage-sensing domain affect the alternative ion permeation pathway in the TRPM3 channel. J Physiol 2018; 596:2413-2432. [PMID: 29604058 PMCID: PMC6002228 DOI: 10.1113/jp274124] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/19/2018] [Indexed: 01/23/2023] Open
Abstract
Key points Mutagenesis at positively charged amino acids (arginines and lysines) (R1–R4) in the voltage‐sensor domain (transmembrane segment (S) 4) of voltage‐gated Na+, K+ and Ca2+ channels can lead to an alternative ion permeation pathway distinct from the central pore. Recently, a non‐canonical ion permeation pathway was described in TRPM3, a member of the transient receptor potential (TRP) superfamily. The non‐canonical pore exists in the native TRPM3 channel and can be activated by co‐stimulation of the endogenous agonist pregnenolone sulphate and the antifungal drug clotrimazole or by stimulation of the synthetic agonist CIM0216. Alignment of the voltage sensor of Shaker K+ channels with the entire TRPM3 sequence revealed the highest degree of similarity in the putative S4 region of TRPM3, and suggested that only one single gating charge arginine (R2) in the putative S4 region is conserved. Mutagenesis studies in the voltage‐sensing domain of TRPM3 revealed several residues in the voltage sensor (S4) as well as in S1 and S3 that are crucial for the occurrence of the non‐canonical inward currents. In conclusion, this study provides evidence for the involvement of the voltage‐sensing domain of TRPM3 in the formation of an alternative ion permeation pathway.
Abstract Transient receptor potential (TRP) channels are cationic channels involved in a broad array of functions, including homeostasis, motility and sensory functions. TRP channel subunits consist of six transmembrane segments (S1–S6), and form tetrameric channels with a central pore formed by the region encompassing S5 and S6. Recently, evidence was provided for the existence of an alternative ion permeation pathway in TRPM3, which allows large inward currents upon hyperpolarization independently of the central pore. However, very little knowledge is available concerning the localization of this alternative pathway in the native TRPM3 channel protein. Guided by sequence homology with Shaker K+ channels, in which mutations in S4 can create an analogous ‘omega’ pore, we performed site‐directed mutagenesis studies and patch clamp experiments to identify amino acid residues involved in the formation of the non‐canonical pore in TRPM3. Based on our results, we pinpoint four residues in S4 (W982, R985, D988 and G991) as crucial determinants of the properties of the alternative ion permeation pathway. Mutagenesis at positively charged amino acids (arginines and lysines) (R1–R4) in the voltage‐sensor domain (transmembrane segment (S) 4) of voltage‐gated Na+, K+ and Ca2+ channels can lead to an alternative ion permeation pathway distinct from the central pore. Recently, a non‐canonical ion permeation pathway was described in TRPM3, a member of the transient receptor potential (TRP) superfamily. The non‐canonical pore exists in the native TRPM3 channel and can be activated by co‐stimulation of the endogenous agonist pregnenolone sulphate and the antifungal drug clotrimazole or by stimulation of the synthetic agonist CIM0216. Alignment of the voltage sensor of Shaker K+ channels with the entire TRPM3 sequence revealed the highest degree of similarity in the putative S4 region of TRPM3, and suggested that only one single gating charge arginine (R2) in the putative S4 region is conserved. Mutagenesis studies in the voltage‐sensing domain of TRPM3 revealed several residues in the voltage sensor (S4) as well as in S1 and S3 that are crucial for the occurrence of the non‐canonical inward currents. In conclusion, this study provides evidence for the involvement of the voltage‐sensing domain of TRPM3 in the formation of an alternative ion permeation pathway.
Collapse
Affiliation(s)
- Katharina Held
- Laboratory of Experimental Gynecology and G-PURE, KU Leuven, Department of Development and Regeneration, Herestraat 49 box 611, B-3000, Leuven, Belgium.,Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Department of Cellular and Molecular Medicine, Herestraat 49 box 802, B-3000, Leuven, Belgium.,VIB Center for Brain & Disease Research, B-3000, Leuven, Belgium
| | - Fabian Gruss
- Laboratory of Structural Neurobiology and TRP Research Platform Leuven (TRPLe), KU Leuven, Department of Cellular and Molecular Medicine, Herestraat 49 box 601, B-3000, Leuven, Belgium
| | - Vincenzo Davide Aloi
- Laboratory of Experimental Gynecology and G-PURE, KU Leuven, Department of Development and Regeneration, Herestraat 49 box 611, B-3000, Leuven, Belgium.,Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Department of Cellular and Molecular Medicine, Herestraat 49 box 802, B-3000, Leuven, Belgium.,VIB Center for Brain & Disease Research, B-3000, Leuven, Belgium
| | - Annelies Janssens
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Department of Cellular and Molecular Medicine, Herestraat 49 box 802, B-3000, Leuven, Belgium.,VIB Center for Brain & Disease Research, B-3000, Leuven, Belgium
| | - Chris Ulens
- Laboratory of Structural Neurobiology and TRP Research Platform Leuven (TRPLe), KU Leuven, Department of Cellular and Molecular Medicine, Herestraat 49 box 601, B-3000, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Department of Cellular and Molecular Medicine, Herestraat 49 box 802, B-3000, Leuven, Belgium.,VIB Center for Brain & Disease Research, B-3000, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Experimental Gynecology and G-PURE, KU Leuven, Department of Development and Regeneration, Herestraat 49 box 611, B-3000, Leuven, Belgium
| |
Collapse
|
72
|
A historical perspective on the role of sensory nerves in neurogenic inflammation. Semin Immunopathol 2018; 40:229-236. [PMID: 29616309 PMCID: PMC5960476 DOI: 10.1007/s00281-018-0673-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/06/2018] [Indexed: 01/25/2023]
Abstract
The term ‘neurogenic inflammation’ is commonly used, especially with respect to the role of sensory nerves within inflammatory disease. However, despite over a century of research, we remain unclear about the role of these nerves in the vascular biology of inflammation, as compared with their interacting role in pain processing and of their potential for therapeutic manipulation. This chapter attempts to discuss the progress in understanding, from the initial discovery of sensory nerves until the present day. This covers pioneering findings that these nerves exist, are involved in vascular events and act as important sensors of environmental changes, including injury and infection. This is followed by discovery of the contents they release such as the established vasoactive neuropeptides substance P and CGRP as well as anti-inflammatory peptides such as the opioids and somatostatin. The more recent emergence of the importance of the transient receptor potential (TRP) channels has revealed some of the mechanisms by which these nerves sense environmental stimuli. This knowledge enables a platform from which to learn of the potential role of neurogenic inflammation in disease and in turn of novel therapeutic targets.
Collapse
|
73
|
Przibilla J, Dembla S, Rizun O, Lis A, Jung M, Oberwinkler J, Beck A, Philipp SE. Ca 2+-dependent regulation and binding of calmodulin to multiple sites of Transient Receptor Potential Melastatin 3 (TRPM3) ion channels. Cell Calcium 2018; 73:40-52. [PMID: 29880196 DOI: 10.1016/j.ceca.2018.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/19/2018] [Accepted: 03/30/2018] [Indexed: 10/17/2022]
Abstract
TRPM3 proteins assemble to Ca2+-permeable cation channels in the plasma membrane, which act as nociceptors of noxious heat and mediators of insulin and cytokine release. Here we show that TRPM3 channel activity is strongly dependent on intracellular Ca2+. Conceivably, this effect is attributed to the Ca2+ binding protein calmodulin, which binds to TRPM3 in a Ca2+-dependent manner. We identified five calmodulin binding sites within the amino terminus of TRPM3, which displayed different binding affinities in dependence of Ca2+. Mutations of lysine residues in calmodulin binding site 2 strongly reduced calmodulin binding and TRPM3 activity indicating the importance of this domain for TRPM3-mediated Ca2+ signaling. Our data show that TRPM3 channels are regulated by intracellular Ca2+ and provide the basis for a mechanistic understanding of the regulation of TRPM3 by calmodulin.
Collapse
Affiliation(s)
- Julia Przibilla
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Sandeep Dembla
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Oleksandr Rizun
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Annette Lis
- Department of Biophysics, Centre for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg 66421, Germany
| | - Martin Jung
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Johannes Oberwinkler
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, 35037 Marburg, Germany
| | - Andreas Beck
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany; Zentrum für Human- und Molekularbiologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Stephan E Philipp
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany.
| |
Collapse
|
74
|
Moore C, Gupta R, Jordt SE, Chen Y, Liedtke WB. Regulation of Pain and Itch by TRP Channels. Neurosci Bull 2018; 34:120-142. [PMID: 29282613 PMCID: PMC5799130 DOI: 10.1007/s12264-017-0200-8] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023] Open
Abstract
Nociception is an important physiological process that detects harmful signals and results in pain perception. In this review, we discuss important experimental evidence involving some TRP ion channels as molecular sensors of chemical, thermal, and mechanical noxious stimuli to evoke the pain and itch sensations. Among them are the TRPA1 channel, members of the vanilloid subfamily (TRPV1, TRPV3, and TRPV4), and finally members of the melastatin group (TRPM2, TRPM3, and TRPM8). Given that pain and itch are pro-survival, evolutionarily-honed protective mechanisms, care has to be exercised when developing inhibitory/modulatory compounds targeting specific pain/itch-TRPs so that physiological protective mechanisms are not disabled to a degree that stimulus-mediated injury can occur. Such events have impeded the development of safe and effective TRPV1-modulating compounds and have diverted substantial resources. A beneficial outcome can be readily accomplished via simple dosing strategies, and also by incorporating medicinal chemistry design features during compound design and synthesis. Beyond clinical use, where compounds that target more than one channel might have a place and possibly have advantageous features, highly specific and high-potency compounds will be helpful in mechanistic discovery at the structure-function level.
Collapse
Affiliation(s)
- Carlene Moore
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Rupali Gupta
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yong Chen
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Wolfgang B Liedtke
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
75
|
Abstract
Heat sensation, the ability to detect warm and noxious temperatures, is an ancient and indispensable sensory process. Noxious temperatures can have detrimental effects on the physiology and integrity of cells, and therefore, the detection of environmental hot temperatures is absolutely crucial for survival. Temperature-sensitive ion channels, which conduct ions in a highly temperature-dependent manner, have been put forward as molecular thermometers expressed at the endings of sensory neurons. In particular, several temperature-sensitive members of the transient receptor potential (TRP) superfamily of ion channels have been identified, and a multitude of in vivo studies have shown that the capsaicin-sensitive TRPV1 channel plays a key role as a noxious heat sensor. However, Trpv1-deficient mice display a residual heat sensitivity suggesting the existence of additional heat sensor(s). In this chapter, we provide evidence for the role of the non-selective calcium-permeable TRPM3 ion channel as an additional heat sensor that acts independently of TRPV1, and give an update of the modulation of this channel by various molecular mechanisms. Finally, we compare antagonists of TRPM3 to specific blockers of TRPV1 as potential analgesic drugs to treat pathological pain.
Collapse
|
76
|
Abstract
The sensation of pain plays a vital protecting role, alerting organisms about potentially damaging stimuli. Tissue injury is detected by nerve endings of specialized peripheral sensory neurons called nociceptors that are equipped with different ion channels activated by thermal, mechanic, and chemical stimuli. Several transient receptor potential channels have been identified as molecular transducers of thermal stimuli in pain-sensing neurons. Skin injury or inflammation leads to increased sensitivity to thermal and mechanic stimuli, clinically defined as allodynia or hyperalgesia. This hypersensitivity is also characteristic of systemic inflammatory disorders and neuropathic pain conditions. Mechanisms of thermal hyperalgesia include peripheral sensitization of nociceptor afferents and maladaptive changes in pain-encoding neurons within the central nervous system. An important aspect of pain management involves attempts to minimize the development of nociceptor hypersensitivity. However, knowledge about the cellular and molecular mechanisms causing thermal hyperalgesia and allodynia in human subjects is still limited, and such knowledge would be an essential step for the development of more effective therapies.
Collapse
Affiliation(s)
- Félix Viana
- Alicante Institute of Neurosciences, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Spain.
| |
Collapse
|
77
|
Thiel G, Rubil S, Lesch A, Guethlein LA, Rössler OG. Transient receptor potential TRPM3 channels: Pharmacology, signaling, and biological functions. Pharmacol Res 2017; 124:92-99. [DOI: 10.1016/j.phrs.2017.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022]
|
78
|
Thiel G, Lesch A, Rubil S, Backes TM, Rössler OG. Regulation of Gene Transcription Following Stimulation of Transient Receptor Potential (TRP) Channels. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 335:167-189. [PMID: 29305012 DOI: 10.1016/bs.ircmb.2017.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transient receptor potential (TRP) channels belong to a heterogeneous superfamily of cation channels that are involved in the regulation of numerous biological functions, including regulation of Ca2+ and glucose homeostasis, tumorigenesis, temperature, and pain sensation. To understand the functions of TRP channels, their associated intracellular signaling pathways and molecular targets have to be identified on the cellular level. Stimulation of TRP channels frequently induces an influx of Ca2+ ions into the cells and the subsequent activation of protein kinases. These intracellular signal transduction pathways ultimately induce changes in the gene expression pattern of the cells. Here, we review the effects of TRPC6, TRPM3, and TRPV1 channel stimulation on the activation of the stimulus-responsive transcription factors AP-1, CREB, Egr-1, Elk-1, and NFAT. Following activation, these transcription factors induce the transcription of delayed response genes. We propose that many biological functions of TRP channels can be explained by the activation of stimulus-responsive transcription factors and their delayed response genes. The proteins encoded by those delayed response genes may be responsible for the biochemical and physiological changes following TRP channel activation.
Collapse
Affiliation(s)
- Gerald Thiel
- Saarland University Medical Faculty, Homburg, Germany.
| | - Andrea Lesch
- Saarland University Medical Faculty, Homburg, Germany
| | - Sandra Rubil
- Saarland University Medical Faculty, Homburg, Germany
| | | | | |
Collapse
|
79
|
Badheka D, Yudin Y, Borbiro I, Hartle CM, Yazici A, Mirshahi T, Rohacs T. Inhibition of Transient Receptor Potential Melastatin 3 ion channels by G-protein βγ subunits. eLife 2017; 6. [PMID: 28829742 PMCID: PMC5593506 DOI: 10.7554/elife.26147] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/28/2017] [Indexed: 11/26/2022] Open
Abstract
Transient receptor potential melastatin 3 (TRPM3) channels are activated by heat, and chemical ligands such as pregnenolone sulphate (PregS) and CIM0216. Here, we show that activation of receptors coupled to heterotrimeric Gi/o proteins inhibits TRPM3 channels. This inhibition was alleviated by co-expression of proteins that bind the βγ subunits of heterotrimeric G-proteins (Gβγ). Co-expression of Gβγ, but not constitutively active Gαi or Gαo, inhibited TRPM3 currents. TRPM3 co-immunoprecipitated with Gβ, and purified Gβγ proteins applied to excised inside-out patches inhibited TRPM3 currents, indicating a direct effect. Baclofen and somatostatin, agonists of Gi-coupled receptors, inhibited Ca2+ signals induced by PregS and CIM0216 in mouse dorsal root ganglion (DRG) neurons. The GABAB receptor agonist baclofen also inhibited inward currents induced by CIM0216 in DRG neurons, and nocifensive responses elicited by this TRPM3 agonist in mice. Our data uncover a novel signaling mechanism regulating TRPM3 channels. DOI:http://dx.doi.org/10.7554/eLife.26147.001
Collapse
Affiliation(s)
- Doreen Badheka
- New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, United States
| | - Yevgen Yudin
- New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, United States
| | - Istvan Borbiro
- New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, United States
| | - Cassandra M Hartle
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Aysenur Yazici
- New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, United States
| | - Tooraj Mirshahi
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Tibor Rohacs
- New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, United States
| |
Collapse
|
80
|
Quallo T, Alkhatib O, Gentry C, Andersson DA, Bevan S. G protein βγ subunits inhibit TRPM3 ion channels in sensory neurons. eLife 2017; 6. [PMID: 28826490 PMCID: PMC5593501 DOI: 10.7554/elife.26138] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) ion channels in peripheral sensory neurons are functionally regulated by hydrolysis of the phosphoinositide PI(4,5)P2 and changes in the level of protein kinase mediated phosphorylation following activation of various G protein coupled receptors. We now show that the activity of TRPM3 expressed in mouse dorsal root ganglion (DRG) neurons is inhibited by agonists of the Gi-coupled µ opioid, GABA-B and NPY receptors. These agonist effects are mediated by direct inhibition of TRPM3 by Gβγ subunits, rather than by a canonical cAMP mediated mechanism. The activity of TRPM3 in DRG neurons is also negatively modulated by tonic, constitutive GPCR activity as TRPM3 responses can be potentiated by GPCR inverse agonists. GPCR regulation of TRPM3 is also seen in vivo where Gi/o GPCRs agonists inhibited and inverse agonists potentiated TRPM3 mediated nociceptive behavioural responses. DOI:http://dx.doi.org/10.7554/eLife.26138.001 TRPM3 belongs to a family of channel proteins that allow sodium and calcium ions to enter cells by forming pores in cell membranes. TRPM3 is found on the cell membranes of nerve cells; when ions flow into the nerves through the TRPM3 pores it triggers an electrical impulse. TRPM3 is responsible for helping us to detect heat, and mice without this protein find it difficult to sense painfully hot temperatures. Mice lacking TRPM3 also respond to other kinds of pain differently. Normally, a mouse with an injured paw becomes more sensitive to warm and hot temperatures, but this does not happen in mice that do not have TRPM3. When activated, other proteins called G-protein coupled receptors (or GPCRs for short) can make some members of this family of channel proteins more or less likely to open their pore. This in turn increases or decreases the flow of ions through the pore, respectively. Yet it was not clear if GPCRs also affect TRPM3 channels on the membranes of nerve cells. Quallo et al. have now discovered that “switching on” different GPCR proteins in sensory nerve cells from mice greatly reduces the flow of calcium ions though TRPM3 channels. The experiments made use of two pain-killing drugs, namely morphine and baclofen, and a molecule called neuropeptide Y to activate different GPCRs. GPCRs interact with a group of small proteins called G-proteins that, when activated by the receptor, split into two subunits, known as the α subunit and the βγ subunit. Once detached these subunits are free to act as messengers and interact with other proteins in the cell membrane. Quallo et al. found that TRPM3 is one of a small group of proteins that interact with the βγ subunits of the G-protein, which can explain how “switching on” GPCRs reduces the activity of TRPM3. Two independent studies by Dembla, Behrendt et al. and Badheka, Yudin et al. also report similar findings. There is currently a need to find more effective treatments for people suffering from long-term pain conditions and it has become clear that TRPM3 channels are involved in sensing both pain and temperature. These new findings show that drugs already used in the treatment of pain can dramatically change how TRPM3 works. These results might help scientists to find drugs that work in a similar way to dial down the activity of TRPM3 and to combat pain. Though first it will be important to confirm these new findings in human nerve cells. DOI:http://dx.doi.org/10.7554/eLife.26138.002
Collapse
Affiliation(s)
- Talisia Quallo
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Omar Alkhatib
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Clive Gentry
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - David A Andersson
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Stuart Bevan
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
81
|
Dembla S, Behrendt M, Mohr F, Goecke C, Sondermann J, Schneider FM, Schmidt M, Stab J, Enzeroth R, Leitner MG, Nuñez-Badinez P, Schwenk J, Nürnberg B, Cohen A, Philipp SE, Greffrath W, Bünemann M, Oliver D, Zakharian E, Schmidt M, Oberwinkler J. Anti-nociceptive action of peripheral mu-opioid receptors by G-beta-gamma protein-mediated inhibition of TRPM3 channels. eLife 2017; 6:26280. [PMID: 28826482 PMCID: PMC5593507 DOI: 10.7554/elife.26280] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022] Open
Abstract
Opioids, agonists of µ-opioid receptors (µORs), are the strongest pain killers clinically available. Their action includes a strong central component, which also causes important adverse effects. However, µORs are also found on the peripheral endings of nociceptors and their activation there produces meaningful analgesia. The cellular mechanisms downstream of peripheral µORs are not well understood. Here, we show in neurons of murine dorsal root ganglia that pro-nociceptive TRPM3 channels, present in the peripheral parts of nociceptors, are strongly inhibited by µOR activation, much more than other TRP channels in the same compartment, like TRPV1 and TRPA1. Inhibition of TRPM3 channels occurs via a short signaling cascade involving Gβγ proteins, which form a complex with TRPM3. Accordingly, activation of peripheral µORs in vivo strongly attenuates TRPM3-dependent pain. Our data establish TRPM3 inhibition as important consequence of peripheral µOR activation indicating that pharmacologically antagonizing TRPM3 may be a useful analgesic strategy. There are very few treatments available for people suffering from strong or long-lasting pain. Currently, substances called opioids – which include the well-known drug morphine – are the strongest painkillers. However, these drugs also cause harmful side effects, which makes them less useful. Like all drugs, opioids mediate their effects by interacting with molecules in the body. In the case of opioids, these interacting molecules belong to a group of receptor proteins called G-protein coupled receptors (or GPCRs for short). These opioid receptors are widely distributed in the nerve cells and brain regions that detect and transmit pain signals. It was poorly understood how activation of opioid receptors reduces the activity of pain-sensing nerve cells, however several lines of evidence had suggested that a protein called TRPM3 might be involved. TRPM3 is a channel protein that allows sodium and calcium ions to enter into nerve cells by forming pores in cell membranes, and mice that lack this protein are less sensitive to certain kinds of pain. Dembla, Behrendt et al. now show that activating opioid receptors on nerve cells from mice, with morphine and a similar substance, rapidly reduces the flow of calcium ions through TRPM3 channels. Further experiments confirmed that activating opioid receptors in a mouse’s paw also reduced the pain caused when TRPM3 proteins are activated. GPCRs interact with a group of small proteins called G-proteins that, when activated by the receptor, split into two subunits. Based on studies with human kidney cells, Dembla, Behrendt et al. found the so-called G-beta-gamma subunit then carries the signal from the opioid receptor to TRPM3. Two independent studies by Quallo et al. and Badheka, Yudin et al. also report similar findings. These new findings show that drugs already used in the treatment of pain can indirectly alter how TRPM3 works in a dramatic way. These results might help scientists to find drugs that work in a more direct way to dial down the activity of TRPM3 and to combat pain with fewer side effects. Though first it will be important to confirm these new findings in human nerve cells.
Collapse
Affiliation(s)
- Sandeep Dembla
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Marc Behrendt
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Florian Mohr
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Christian Goecke
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Julia Sondermann
- Max-Planck-Institut für Experimentelle Medizin, Göttingen, Germany
| | - Franziska M Schneider
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Marlene Schmidt
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Julia Stab
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, Germany
| | - Raissa Enzeroth
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Michael G Leitner
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Paulina Nuñez-Badinez
- Department of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim Heidelberg University, Mannheim, Germany
| | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernd Nürnberg
- Abteilung für Pharmakologie und Experimentelle Therapie, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universität Tübingen, Tübingen, Germany
| | - Alejandro Cohen
- Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Stephan E Philipp
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, Germany
| | - Wolfgang Greffrath
- Department of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim Heidelberg University, Mannheim, Germany
| | - Moritz Bünemann
- Institut für Pharmakologie und Klinische Pharmazie, Philipps-Universität Marburg, Marburg, Germany
| | - Dominik Oliver
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Eleonora Zakharian
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, United States
| | - Manuela Schmidt
- Max-Planck-Institut für Experimentelle Medizin, Göttingen, Germany
| | - Johannes Oberwinkler
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
82
|
Belvisi MG, Birrell MA. The emerging role of transient receptor potential channels in chronic lung disease. Eur Respir J 2017; 50:50/2/1601357. [PMID: 28775042 DOI: 10.1183/13993003.01357-2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/14/2017] [Indexed: 12/12/2022]
Abstract
Chronic lung diseases such as asthma, chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis are a major and increasing global health burden with a high unmet need. Drug discovery efforts in this area have been largely disappointing and so new therapeutic targets are needed. Transient receptor potential ion channels are emerging as possible therapeutic targets, given their widespread expression in the lung, their role in the modulation of inflammatory and structural changes and in the production of respiratory symptoms, such as bronchospasm and cough, seen in chronic lung disease.
Collapse
Affiliation(s)
- Maria G Belvisi
- Respiratory Pharmacology Group, Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Mark A Birrell
- Respiratory Pharmacology Group, Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
83
|
Jia S, Zhang Y, Yu J. Antinociceptive Effects of Isosakuranetin in a Rat Model of Peripheral Neuropathy. Pharmacology 2017; 100:201-207. [PMID: 28715803 DOI: 10.1159/000478986] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/26/2017] [Indexed: 12/27/2022]
Abstract
Chronic pain remains a challenging clinical reality, yet currently available analgesics are insufficient to meet clinical needs. Increasing attention has been paid to bioactive compounds from natural plants, which may be efficacious against pain. This study examined the antinociceptive effects of isosakuranetin, a plant-derived transient receptor potential melastatin 3 blocker, in a rat model of peripheral neuropathy. Adult male Sprague-Dawley rats were first allowed to go through the chronic constriction injury surgery to develop neuropathic pain. They were then treated with isosakuranetin (1.5, 3, or 6 mg/kg) intraperitoneally and the effects on mechanical, thermal, and cold hyperalgesia were assessed using the von Frey filament test, Hargreaves' plantar test, and cold plate test, respectively. Isosakuranetin dose-dependently alleviated mechanical, thermal, and cold hyperalgesia and the antinociceptive potency was similar across the assays. In the rotarod test, isosakuranetin did not significantly affect motor performance within the doses tested, confirming the antinociceptive specificity. In summary, these findings suggest that isosakuranetin may be useful in treating neuropathic pain and deserves further investigation.
Collapse
Affiliation(s)
- Shushan Jia
- Department of Anesthesiology, Shandong University Qilu Hospital, Jinan, China
| | | | | |
Collapse
|
84
|
Malenczyk K, Girach F, Szodorai E, Storm P, Segerstolpe Å, Tortoriello G, Schnell R, Mulder J, Romanov RA, Borók E, Piscitelli F, Di Marzo V, Szabó G, Sandberg R, Kubicek S, Lubec G, Hökfelt T, Wagner L, Groop L, Harkany T. A TRPV1-to-secretagogin regulatory axis controls pancreatic β-cell survival by modulating protein turnover. EMBO J 2017; 36:2107-2125. [PMID: 28637794 PMCID: PMC5510001 DOI: 10.15252/embj.201695347] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 04/27/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022] Open
Abstract
Ca2+-sensor proteins are generally implicated in insulin release through SNARE interactions. Here, secretagogin, whose expression in human pancreatic islets correlates with their insulin content and the incidence of type 2 diabetes, is shown to orchestrate an unexpectedly distinct mechanism. Single-cell RNA-seq reveals retained expression of the TRP family members in β-cells from diabetic donors. Amongst these, pharmacological probing identifies Ca2+-permeable transient receptor potential vanilloid type 1 channels (TRPV1) as potent inducers of secretagogin expression through recruitment of Sp1 transcription factors. Accordingly, agonist stimulation of TRPV1s fails to rescue insulin release from pancreatic islets of glucose intolerant secretagogin knock-out(-/-) mice. However, instead of merely impinging on the SNARE machinery, reduced insulin availability in secretagogin-/- mice is due to β-cell loss, which is underpinned by the collapse of protein folding and deregulation of secretagogin-dependent USP9X deubiquitinase activity. Therefore, and considering the desensitization of TRPV1s in diabetic pancreata, a TRPV1-to-secretagogin regulatory axis seems critical to maintain the structural integrity and signal competence of β-cells.
Collapse
Affiliation(s)
- Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fatima Girach
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Petter Storm
- Department of Clinical Sciences, Diabetes and Endocrinology CRC, Skåne University Hospital Malmö, Malmö, Sweden
| | - Åsa Segerstolpe
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | | | - Robert Schnell
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jan Mulder
- Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Erzsébet Borók
- Department of Cognitive Neurobiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli Naples, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli Naples, Italy
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Rickard Sandberg
- Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | - Stefan Kubicek
- CeMM Research Centre for Molecular Medicine, Vienna, Austria
| | - Gert Lubec
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ludwig Wagner
- University Clinic for Internal Medicine III, General Hospital Vienna, Vienna, Austria
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology CRC, Skåne University Hospital Malmö, Malmö, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
85
|
Abstract
Supplemental Digital Content is Available in the Text. The approved antiepileptic drug primidone potently inhibits TRPM3 channels and thereby exerts analgesic properties to chemical pain and thermal hyperalgesia in mice. The melastatin-related transient receptor potential (TRP) channel TRPM3 is a nonselective cation channel expressed in nociceptive neurons and activated by heat. Because TRPM3-deficient mice show inflammatory thermal hyperalgesia, pharmacological inhibition of TRPM3 may exert antinociceptive properties. Fluorometric Ca2+ influx assays and a compound library containing approved or clinically tested drugs were used to identify TRPM3 inhibitors. Biophysical properties of channel inhibition were assessed using electrophysiological methods. The nonsteroidal anti-inflammatory drug diclofenac, the tetracyclic antidepressant maprotiline, and the anticonvulsant primidone were identified as highly efficient TRPM3 blockers with half-maximal inhibition at 0.6 to 6 μM and marked specificity for TRPM3. Most prominently, primidone was biologically active to suppress TRPM3 activation by pregnenolone sulfate (PregS) and heat at concentrations markedly lower than plasma concentrations commonly used in antiepileptic therapy. Primidone blocked PregS-induced Ca2+i influx through TRPM3 by allosteric modulation and reversibly inhibited atypical inwardly rectifying TRPM3 currents induced by coapplication of PregS and clotrimazole. In vivo, analgesic effects of low doses of primidone were demonstrated in mice, applying PregS- and heat-induced pain models, including inflammatory hyperalgesia. Thus, applying the approved drug at concentrations that are lower than those needed to induce anticonvulsive effects offers a shortcut for studying physiological and pathophysiological roles of TRPM3 in vivo.
Collapse
|
86
|
Helås T, Sagafos D, Kleggetveit I, Quiding H, Jönsson B, Segerdahl M, Zhang Z, Salter H, Schmelz M, Jørum E. Pain thresholds,supra-threshold pain and lidocaine sensitivity in patients with erythromelalgia, including the I848Tmutation in NaV1.7. Eur J Pain 2017; 21:1316-1325. [DOI: 10.1002/ejp.1030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2017] [Indexed: 11/09/2022]
Affiliation(s)
- T. Helås
- Section of Clinical Neurophysiology, Department of Neurology; Oslo University Hospital - Rikshospitalet; Norway
| | - D. Sagafos
- Section of Clinical Neurophysiology, Department of Neurology; Oslo University Hospital - Rikshospitalet; Norway
| | - I.P. Kleggetveit
- Section of Clinical Neurophysiology, Department of Neurology; Oslo University Hospital - Rikshospitalet; Norway
| | | | | | | | - Z. Zhang
- Astra-Zeneca R&D; Södertälje Sweden
| | - H. Salter
- Astra-Zeneca R&D; Södertälje Sweden
- Department of Clinical Neuroscience; Karolinska Institutet; Solna Sweden
| | - M. Schmelz
- Department of Anesthesiology Mannheim; Heidelberg University; Germany
| | - E. Jørum
- Section of Clinical Neurophysiology, Department of Neurology; Oslo University Hospital - Rikshospitalet; Norway
- Faculty of Medicine, Institute of Clinical Medicine; University of Oslo; Norway
| |
Collapse
|
87
|
Siroky BJ, Kleene NK, Kleene SJ, Varnell CD, Comer RG, Liu J, Lu L, Pachciarz NW, Bissler JJ, Dixon BP. Primary cilia regulate the osmotic stress response of renal epithelial cells through TRPM3. Am J Physiol Renal Physiol 2017; 312:F791-F805. [PMID: 28122715 PMCID: PMC5407065 DOI: 10.1152/ajprenal.00465.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 12/26/2022] Open
Abstract
Primary cilia sense environmental conditions, including osmolality, but whether cilia participate in the osmotic response in renal epithelial cells is not known. The transient receptor potential (TRP) channels TRPV4 and TRPM3 are osmoresponsive. TRPV4 localizes to cilia in certain cell types, while renal subcellular localization of TRPM3 is not known. We hypothesized that primary cilia are required for maximal activation of the osmotic response of renal epithelial cells and that ciliary TRPM3 and TRPV4 mediate that response. Ciliated [murine epithelial cells from the renal inner medullary collecting duct (mIMCD-3) and 176-5] and nonciliated (176-5Δ) renal cells expressed Trpv4 and Trpm3 Ciliary expression of TRPM3 was observed in mIMCD-3 and 176-5 cells and in wild-type mouse kidney tissue. TRPV4 was identified in cilia and apical membrane of mIMCD-3 cells by electrophysiology and in the cell body by immunofluorescence. Hyperosmolal stress at 500 mOsm/kg (via NaCl addition) induced the osmotic response genes betaine/GABA transporter (Bgt1) and aldose reductase (Akr1b3) in all ciliated cell lines. This induction was attenuated in nonciliated cells. A TRPV4 agonist abrogated Bgt1 and Akr1b3 induction in ciliated and nonciliated cells. A TRPM3 agonist attenuated Bgt1 and Akr1b3 induction in ciliated cells only. TRPM3 knockout attenuated Akr1b3 induction. Viability under osmotic stress was greater in ciliated than nonciliated cells. Akr1b3 induction was also less in nonciliated than ciliated cells when mannitol was used to induce hyperosmolal stress. These findings suggest that primary cilia are required for the maximal osmotic response in renal epithelial cells and that TRPM3 is involved in this mechanism. TRPV4 appears to modulate the osmotic response independent of cilia.
Collapse
Affiliation(s)
- Brian J Siroky
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nancy K Kleene
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio; and
| | - Steven J Kleene
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio; and
| | - Charles D Varnell
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Raven G Comer
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jialiu Liu
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lu Lu
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nolan W Pachciarz
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John J Bissler
- St. Jude Children's Research Hospital and Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Bradley P Dixon
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| |
Collapse
|
88
|
Mack GW, Foote KM, Nelson WB. Cutaneous Vasodilation during Local Heating: Role of Local Cutaneous Thermosensation. Front Physiol 2016; 7:622. [PMID: 28066257 PMCID: PMC5167758 DOI: 10.3389/fphys.2016.00622] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/28/2016] [Indexed: 11/13/2022] Open
Abstract
We tested the hypothesis that cutaneous vasodilation during local skin heating in humans could be manipulated based upon the ability to desensitize TRPV4 ion channels by applying the thermal stimuli in a series of pulses. Each subject was instrumented with intradermal microdialysis probes in the dorsal forearm skin and perfused with 0.9% saline at 1.5 μl/min with local skin temperature controlled with a Peltier unit (9 cm2) at 34°C. Local skin temperature was manipulated for 50 min in two classic ways: a step increase to 38°C (0.1°C/s, n = 10), and a step increase to 42°C (n = 10). To desensitize TRPV4 ion channels local skin temperature was manipulated in the following way: pulsed increase to 38°C (1 pulse per min, 30 s duration, 1.0°C/s, n = 10), and 4) pulsed increase to 42°C (1.0°C/s, n = 9). Skin blood flow (SkBF, laser Doppler) was recorded directly over the middle microdialysis probe and the dialysate from all three probes were collected during baseline (34°C) and each skin heating period. The overall cutaneous vascular conductance (CVC) response to local heating was estimated from the area under the % CVCmax-time curve. The appearance of the neuropeptide calcitonin gene related peptide (CGRP) in dialysate did not change with skin heating in any protocol. For the skin temperature challenge of 34 to 38°C, the area under the % CVCmax-time curve averaged 1196 ± 295 (SD) % CVCmax•min, which was larger than the 656 ± 282% CVCmax•min during pulsed heating (p < 0.05). For the skin temperature challenge of 34 to 42°C, the area under the % CVCmax-time curve averaged 2678 ± 458% CVCmax•min, which was larger than the 1954 ± 533% CVCmax•min during pulsed heating (p < 0.05). The area under the % CVCmax•min curve, was directly proportional to the accumulated local skin thermal stress (in °C•min) (r2 = 0.62, p < 0.05, n = 39). This association indicates a critical role of local integration of thermosensitive receptors in mediating the cutaneous vasodilator response to local skin heating. Given that we saw no differences in the levels of CGRP in dialysate, the role of the vasoactive peptide CGRP in the cutaneous vasodilator response to local skin heating is not supported by our data.
Collapse
Affiliation(s)
- Gary W Mack
- Department of Exercise Sciences, The Human Performance Research Center, Brigham Young University Provo, UT, USA
| | - Kristopher M Foote
- Department of Anesthesiology, 1500 E Medical Center Drive, University of Michigan Ann Arbor, MI, USA
| | - W Bradley Nelson
- Department of Natural Sciences, Ohio Dominican University Columbus, OH, USA
| |
Collapse
|
89
|
TRP Channels as Therapeutic Targets in Diabetes and Obesity. Pharmaceuticals (Basel) 2016; 9:ph9030050. [PMID: 27548188 PMCID: PMC5039503 DOI: 10.3390/ph9030050] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/16/2022] Open
Abstract
During the last three to four decades the prevalence of obesity and diabetes mellitus has greatly increased worldwide, including in the United States. Both the short- and long-term forecasts predict serious consequences for the near future, and encourage the development of solutions for the prevention and management of obesity and diabetes mellitus. Transient receptor potential (TRP) channels were identified in tissues and organs important for the control of whole body metabolism. A variety of TRP channels has been shown to play a role in the regulation of hormone release, energy expenditure, pancreatic function, and neurotransmitter release in control, obese and/or diabetic conditions. Moreover, dietary supplementation of natural ligands of TRP channels has been shown to have potential beneficial effects in obese and diabetic conditions. These findings raised the interest and likelihood for potential drug development. In this mini-review, we discuss possibilities for better management of obesity and diabetes mellitus based on TRP-dependent mechanisms.
Collapse
|
90
|
Rubil S, Thiel G. Activation of gene transcription via CIM0216, a synthetic ligand of transient receptor potential melastatin-3 (TRPM3) channels. Channels (Austin) 2016; 11:79-83. [PMID: 27356187 DOI: 10.1080/19336950.2016.1207026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Several compounds have been proposed to stimulate TRPM3 Ca2+ channels. We recently showed that stimulation of TRPM3 channels with pregnenolone sulfate activated the transcription factor AP-1, while other proposed TRPM3 ligands (nifedipine, D-erythro-sphingosine) exhibited either no or TRPM3-independent effects on gene transcription. Here, we have analyzed the transcriptional activity of CIM0216, a synthetic TRPM3 ligand proposed to have a higher potency and affinity for TRPM3 than pregnenolone sulfate. The results show that CIM0216 treatment of HEK293 cells expressing TRPM3 channels activated AP-1 and stimulated the transcriptional activation potential of c-Jun and c-Fos, 2 basic region leucine zipper transcription factors that constitute AP-1. CIM0216-induced gene transcription was attenuated by knock-down of TRPM3 or treatment with mefenamic acid, a TRPM3 inhibitor. CIM0216 was similarly or less capable in activating TRPM3-mediated gene transcription, suggesting that pregnenolone sulfate is still the ligand of choice for changing the gene expression pattern via TRPM3.
Collapse
Affiliation(s)
- Sandra Rubil
- a Department of Medical Biochemistry and Molecular Biology , Saarland University Medical Faculty , Homburg , Germany
| | - Gerald Thiel
- a Department of Medical Biochemistry and Molecular Biology , Saarland University Medical Faculty , Homburg , Germany
| |
Collapse
|
91
|
Kichko TI, Pfirrmann RW, Reeh PW. Taurolidine and congeners activate hTRPA1 but not hTRPV1 channels and stimulate CGRP release from mouse tracheal sensory nerves. Pharmacol Res Perspect 2016; 4:e00204. [PMID: 26977296 PMCID: PMC4777271 DOI: 10.1002/prp2.204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/12/2022] Open
Abstract
Taurolidine has long been in clinical use as an antimicrobial irrigation that does not impede wound healing. It can even be administered intravenously (30 g/day) to treat sepsis or to exert newly recognized antineoplastic actions. Only one irritant effect is reported, that is, to temporarily induce burning pain of unknown origin when applied to body cavities or peripheral veins. The structure of the molecule suggested the chemoreceptor channel TRPA1 as a potential target, which was verified measuring stimulated CGRP release from sensory nerves of the isolated mouse trachea and calcium influx in hTRPA1‐transfected HEK293 cells. With both methods, the concentration–response relationship of taurolidine exceeded the threshold value below 500 μmol/L and 100 μmol/L, respectively, and reached saturation at 1 mmol/L. The clinical 2% taurolidine solution did not evoke greater or longer lasting responses. The reversible tracheal response was abolished in TRPA1−/− but retained in TRPV1−/− mice. Consistently, hTRPV1‐HEK showed no calcium influx as a response, likewise native HEK293 cells and hTRPA1‐HEK deprived of extracellular calcium did not respond to taurolidine 1 mmol/L. The metabolite taurultam and its oxathiazine derivative, expected to cause less burning pain, showed weak tracheal irritancy only at 10 mmol/L, acting also through hTRPA1 but not hTRPV1. In conclusion, taurolidine, its metabolite, and a novel derivative showed no unspecific cellular effects but selectively, concentration‐dependently and reversibly activated the irritant receptor TRPA1 in CGRP‐expressing, thus nociceptive, neurons. The clinical solution of 2% taurolidine (~70 mmol/L) can, thus, rightly be expected to cause transient burning pain and neurogenic inflammation.
Collapse
Affiliation(s)
- Tatjana I Kichko
- Institute of Physiology and Pathophysiology Friedrich-Alexander-University of Erlangen-Nürnberg Erlangen Germany
| | | | - Peter W Reeh
- Institute of Physiology and Pathophysiology Friedrich-Alexander-University of Erlangen-Nürnberg Erlangen Germany
| |
Collapse
|
92
|
Held K, Voets T, Vriens J. Signature and Pathophysiology of Non-canonical Pores in Voltage-Dependent Cation Channels. Rev Physiol Biochem Pharmacol 2016; 170:67-99. [DOI: 10.1007/112_2015_5003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
93
|
Uchida K, Demirkhanyan L, Asuthkar S, Cohen A, Tominaga M, Zakharian E. Stimulation-dependent gating of TRPM3 channel in planar lipid bilayers. FASEB J 2015; 30:1306-16. [PMID: 26655382 DOI: 10.1096/fj.15-281576] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022]
Abstract
The transient receptor potential melastatin (TRPM)-3 channel is critical for various physiologic processes. In somatosensory neurons, TRPM3 has been implicated in temperature perception and inflammatory hyperalgesia, whereas in pancreatic β-cells the channel has been linked to glucose-induced insulin release. As a typical representative of the TRP family, TRPM3 is highly polymodal. In cells, it is activated by heat and chemical agonists, including pregnenolone sulfate (PS) and nifedipine (Nif). To define the nuances of TRPM3 channel activity and its modulators, we succeeded in incorporating the TRPM3 protein into planar lipid bilayers. We found that phosphatidylinositol-4,5-bisphosphate (PIP2) or clotrimazole is necessary for channel opening by PS. Unlike PS, the presence of Nif alone sufficed to induce TRPM3 activity and demonstrated distinct gating behavior. We also performed an extensive thermodynamic analysis of TRPM3 activation and found that TRPM3 exhibited slight temperature sensitivity in the bilayers. In the absence of other agonists TRPM3 channels remained closed upon heat-induced stimulation, but opened in the presence of PIP2, although with only a low open-probability profile. Together, our results elucidate the details peculiar to TRPM3 channel function in an isolated system. We confirmed its direct gating by PS and PIP2, but found a lack of the strong intrinsic temperature sensitivity common to other thermosensitive TRP channels.
Collapse
Affiliation(s)
- Kunitoshi Uchida
- *Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA; Division of Cell Signaling, National Institute of Physiological Sciences, Okazaki Institute of Integrative Bioscience, Aichi, Japan; Department of Physiological Sciences, The Graduate University of Advanced Studies, Kanagawa, Japan; and Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lusine Demirkhanyan
- *Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA; Division of Cell Signaling, National Institute of Physiological Sciences, Okazaki Institute of Integrative Bioscience, Aichi, Japan; Department of Physiological Sciences, The Graduate University of Advanced Studies, Kanagawa, Japan; and Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Swapna Asuthkar
- *Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA; Division of Cell Signaling, National Institute of Physiological Sciences, Okazaki Institute of Integrative Bioscience, Aichi, Japan; Department of Physiological Sciences, The Graduate University of Advanced Studies, Kanagawa, Japan; and Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alejandro Cohen
- *Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA; Division of Cell Signaling, National Institute of Physiological Sciences, Okazaki Institute of Integrative Bioscience, Aichi, Japan; Department of Physiological Sciences, The Graduate University of Advanced Studies, Kanagawa, Japan; and Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Makoto Tominaga
- *Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA; Division of Cell Signaling, National Institute of Physiological Sciences, Okazaki Institute of Integrative Bioscience, Aichi, Japan; Department of Physiological Sciences, The Graduate University of Advanced Studies, Kanagawa, Japan; and Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Eleonora Zakharian
- *Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois, USA; Division of Cell Signaling, National Institute of Physiological Sciences, Okazaki Institute of Integrative Bioscience, Aichi, Japan; Department of Physiological Sciences, The Graduate University of Advanced Studies, Kanagawa, Japan; and Proteomics and Mass Spectrometry Core Facility, Life Sciences Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
94
|
Quallo T, Gentry C, Bevan S, Broad LM, Mogg AJ. Activation of transient receptor potential ankyrin 1 induces CGRP release from spinal cord synaptosomes. Pharmacol Res Perspect 2015; 3:e00191. [PMID: 27022465 PMCID: PMC4777244 DOI: 10.1002/prp2.191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/02/2015] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is a sensor of nociceptive stimuli, expressed predominantly in a subpopulation of peptidergic sensory neurons which co-express the noxious heat-sensor transient receptor potential vanilloid 1. In this study, we describe a spinal cord synaptosome-calcitonin gene-related peptide (CGRP) release assay for examining activation of TRPA1 natively expressed on the central terminals of dorsal root ganglion neurons. We have shown for the first time that activation of TRPA1 channels expressed on spinal cord synaptosomes by a selection of agonists evokes a concentration-dependent release of CGRP which is inhibited by TRPA1 antagonists. In addition, our results demonstrate that depolarization of spinal cord synaptosomes by a high concentration of KCl induces CGRP release via a T-type calcium channel-dependent mechanism whilst TRPA1-induced CGRP release functions independently of voltage-gated calcium channel activation. Finally, we have shown that pre-treatment of synaptosomes with the opioid agonist, morphine, results in a reduction of depolarization-induced CGRP release. This study has demonstrated the use of a dorsal spinal cord homogenate assay for investigation of natively expressed TRPA1 channels and for modulation of depolarizing stimuli at the level of the dorsal spinal cord.
Collapse
Affiliation(s)
- Talisia Quallo
- Wolfson Centre for Age Related Diseases King's College London London SE1 1UL United Kingdom
| | - Clive Gentry
- Wolfson Centre for Age Related Diseases King's College London London SE1 1UL United Kingdom
| | - Stuart Bevan
- Wolfson Centre for Age Related Diseases King's College London London SE1 1UL United Kingdom
| | - Lisa M Broad
- Neuroscience Research Division Lilly Research Centre Eli Lilly & Co. Ltd Windlesham United Kingdom
| | - Adrian J Mogg
- Neuroscience Research Division Lilly Research Centre Eli Lilly & Co. Ltd Windlesham United Kingdom
| |
Collapse
|