Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Expert Opin Ther Targets ; 28(4): 251-258, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38629152

ABSTRACT

INTRODUCTION: Opinions differ on what drugs have both a rationale and a development potential for the treatment of bladder storage dysfunction. AREAS COVERED: In the present review, the focus is given to small molecule blockers of TRP channels (TRPV1, TRPV4, TRPA1, and TRPM8), P2 × 3receptor antagonists, drugs against oxidative stress, antifibrosis agents, cyclic nucleotide - dependent pathways, and MaxiK±channel - gene therapy. EXPERT OPINION: TRPV1 channel blockers produce hypothermia which seems to be a problem even with the most efficacious second-generation TRPV1 antagonists. This has so far precluded their application to urine storage disorders. Other TRP channel blockers with promising rationale have yet to be tested on the human lower urinary tract. The P2 × 3receptor antagonist, eliapixant, was tested in a randomized controlled clinical trial, was well tolerated but did not meet clinical efficacy endpoints. Antifibrosis agent still await application to the human lower urinary tract. New drug principles for oxidative stress, purine nucleoside phosphorylase inhibition, and NOX inhibition are still at an experimental stage, and so are soluble guanylate cyclase stimulators. Gene therapy with MaxiK±channels is still an interesting approach but no new trials seem to be in pipeline.


Subject(s)
Drug Development , Molecular Targeted Therapy , Oxidative Stress , Humans , Animals , Oxidative Stress/drug effects , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/drug effects , Genetic Therapy/methods , Randomized Controlled Trials as Topic , Urinary Bladder Diseases/drug therapy , Urinary Bladder Diseases/physiopathology
2.
J Nat Prod ; 87(4): 722-732, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38408345

ABSTRACT

The first detailed phytochemical analysis of the cannabigerol (CBG)-rich chemotype IV of Cannabis sativa L. resulted in the isolation of the expected cannabigerolic acid/cannabigerol (CBGA/CBG) and cannabidiolic acid/cannabidiol (CBDA/CBD) and of nine new phytocannabinoids (5-13), which were fully characterized by HR-ESIMS and 1D and 2D NMR. These included mono- or dihydroxylated CBGA/CBG analogues, a congener with a truncated side chain (10), cyclocannabigerol B (11), and the CBD derivatives named cannabifuranols (12 and 13). Cyclocannabigerol B and cannabifuranols are characterized by a novel phytocannabinoid structural architecture. The isolated phytocannabinoids were assayed on the receptor channels TRPA1 and TRPM8, unveiling a potent dual TRPA1 agonist/TRPM8 antagonist profile for compounds 6, 7, and 14. Chiral separation of the two enantiomers of 5 resulted in the discovery of a synergistic effect of the two enantiomers on TRPA1.


Subject(s)
Cannabinoids , Cannabis , TRPA1 Cation Channel , TRPM Cation Channels , Transient Receptor Potential Channels , Cannabis/chemistry , TRPA1 Cation Channel/antagonists & inhibitors , Cannabinoids/pharmacology , Cannabinoids/chemistry , Cannabinoids/isolation & purification , TRPM Cation Channels/antagonists & inhibitors , Molecular Structure , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/drug effects , Phytochemicals/pharmacology , Phytochemicals/isolation & purification , Phytochemicals/chemistry , Humans , Cannabidiol/pharmacology , Cannabidiol/chemistry , Calcium Channels/metabolism
3.
J Biol Chem ; 300(1): 105484, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37992804

ABSTRACT

Sterols are hydrophobic molecules, known to cluster signaling membrane-proteins in lipid rafts, while methyl-ß-cyclodextrin (MßCD) has been a major tool for modulating membrane-sterol content for studying its effect on membrane proteins, including the transient receptor potential (TRP) channels. The Drosophila light-sensitive TRP channels are activated downstream of a G-protein-coupled phospholipase Cß (PLC) cascade. In phototransduction, PLC is an enzyme that hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) generating diacylglycerol, inositol-tris-phosphate, and protons, leading to TRP and TRP-like (TRPL) channel openings. Here, we studied the effects of MßCD on Drosophila phototransduction using electrophysiology while fluorescently monitoring PIP2 hydrolysis, aiming to examine the effects of sterol modulation on PIP2 hydrolysis and the ensuing light-response in the native system. Incubation of photoreceptor cells with MßCD dramatically reduced the amplitude and kinetics of the TRP/TRPL-mediated light response. MßCD also suppressed PLC-dependent TRP/TRPL constitutive channel activity in the dark induced by mitochondrial uncouplers, but PLC-independent activation of the channels by linoleic acid was not affected. Furthermore, MßCD suppressed a constitutively active TRP mutant-channel, trpP365, suggesting that TRP channel activity is a target of MßCD action. Importantly, whole-cell voltage-clamp measurements from photoreceptors and simultaneously monitored PIP2-hydrolysis by translocation of fluorescently tagged Tubby protein domain, from the plasma membrane to the cytosol, revealed that MßCD virtually abolished the light response when having little effect on the light-activated PLC. Together, MßCD uncoupled TRP/TRPL channel gating from light-activated PLC and PIP2-hydrolysis suggesting the involvement of distinct nanoscopic lipid domains such as lipid rafts and PIP2 clusters in TRP/TRPL channel gating.


Subject(s)
Drosophila Proteins , Membrane Lipids , Transient Receptor Potential Channels , Type C Phospholipases , beta-Cyclodextrins , Animals , beta-Cyclodextrins/pharmacology , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Lipids/metabolism , Photoreceptor Cells, Invertebrate/drug effects , Photoreceptor Cells, Invertebrate/metabolism , Sterols/metabolism , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Type C Phospholipases/metabolism , Light Signal Transduction/drug effects
4.
Cancer Lett ; 577: 216435, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37806516

ABSTRACT

Dying tumor cells release biological signals that exhibit antigenicity, activate cytotoxic T lymphocytes, and induce immunogenic cell death (ICD), playing a key role in immune surveillance. We demonstrate that the flavonoid LW-213 activates endoplasmic reticulum stress (ERS) in different tumor cells and that the lysosomal calcium channel TRPML1 mediates the ERS process in human cellular lymphoma Hut-102 cells. Apoptotic tumor cells induced by ERS often possess immunogenicity. Tumor cells treated with LW-213 exhibit damage-associated molecular patterns (DAMPs), including calreticulin translocation to the plasma membrane and extracellular release of ATP and HMGB1. When co-cultured with antigen-presenting cells (APCs), LW-213-treated tumor cells activated APCs. Two groups of C57BL/6J mice were inoculated with Lewis cells: a "vaccine group", which demonstrated that LW-213-treated tumor cells promote the maturation of dendritic cells and increase CD8+ T cells infiltration in the tumor microenvironment and a "pharmacodynamic group", treated with a combination of LW-213 and PD1/PD-L1 inhibitor (BMS-1), which reduced tumor growth and significantly prolonged the survival time of mice in the "pharmacodynamic group". Therefore, LW-213 can be developed as a novel ICD inducer, providing a new concept for antitumor immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Flavonoids , Immunogenic Cell Death , Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Endoplasmic Reticulum Stress , Immunogenic Cell Death/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL , Neoplasms/metabolism , Tumor Microenvironment , Flavonoids/pharmacology , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism
5.
Cell Calcium ; 106: 102640, 2022 09.
Article in English | MEDLINE | ID: mdl-36030694

ABSTRACT

The transient receptor potential cation channel, subfamily M, members 6 and 7 (TRPM6 and TRPM7) are homologous membrane proteins encompassing cation channel units fused to cytosolic serine/threonine-protein kinase domains. Clinical studies and experiments with animal disease models suggested that selective inhibition of TRPM6 and TRPM7 currents might be beneficial for subjects with immune and cardiovascular disorders, tumours and other pathologies, but the suitable pharmacological toolkit remains underdeveloped. The present study identified small synthetic molecules acting specifically on the channel moieties of TRPM6 and TRPM7. Using electrophysiological analysis in conjunction with Ca2+ imaging, we show that iloperidone and ifenprodil inhibit the channel activity of recombinant TRPM6 with IC50 values of 0.73 and 3.33 µM, respectively, without an impact on the TRPM7 channel. We also found that VER155008 suppresses the TRPM7 channel with an IC50 value of 0.11 µM but does not affect TRPM6. Finally, the effects of iloperidone and VER155008 were found to be suitable for blocking native endogenous TRPM6 and TRPM7 in a collection of mouse and human cell models. Hence, the identification of iloperidone, ifenprodil, and VER155008 allows for the first time to selectively manipulate TRPM6 and TRPM7 currents.


Subject(s)
TRPM Cation Channels , Animals , Humans , Isoxazoles/pharmacology , Magnesium/metabolism , Mice , Piperidines/pharmacology , Protein Serine-Threonine Kinases , Purine Nucleosides/pharmacology , TRPM Cation Channels/drug effects , TRPM Cation Channels/metabolism , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism
6.
Sci Rep ; 11(1): 20627, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663887

ABSTRACT

Cnidarians are characterized by the possession of stinging organelles, called nematocysts, which they use for prey capture and defense. Nematocyst discharge is controlled by a mechanosensory apparatus with analogies to vertebrate hair cells. Members of the transient receptor potential (TRPN) ion channel family are supposed to be involved in the transduction of the mechanical stimulus. A small molecule screen was performed to identify compounds that affect nematocyst discharge in Hydra. We identified several [2.2]paracyclophanes that cause inhibition of nematocyst discharge in the low micro-molar range. Further structure-activity analyses within the compound class of [2.2]paracyclophanes showed common features that are required for the inhibitory activity of the [2.2]paracyclophane core motif. This study demonstrates that Hydra can serve as a model for small molecule screens targeting the mechanosensory apparatus in native tissues.


Subject(s)
Hydra/immunology , Nematocyst/drug effects , Nematocyst/physiology , Animals , Biomechanical Phenomena/drug effects , Biomechanical Phenomena/physiology , Cnidaria , Hydra/metabolism , Small Molecule Libraries/pharmacology , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/physiology
7.
Emerg Microbes Infect ; 10(1): 1257-1271, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34102949

ABSTRACT

Japanese encephalitis virus (JEV), a member of the Flavivirus genus, is an important pathogen that causes human and animal infectious diseases in Asia. So far, no effective antiviral agents are available to treat JEV infection. Here, we found that LDLR is a host factor required for JEV entry. Berbamine significantly decreases the level of LDLR at the plasma membrane by inducing the secretion of LDLR via extracellular vesicles (EVs), thereby inhibiting JEV infection. Mechanistically, berbamine blocks TRPMLs (Ca2+ permeable non-selective cation channels in endosomes and lysosomes) to compromise the endolysosomal trafficking of LDLR. This leads to the increased secretion of LDLR via EVs and the concomitant decrease in its level at the plasma membrane, thereby rendering cells resistant to JEV infection. Berbamine also protects mice from the lethal challenge of JEV. In summary, these results indicate that berbamine is an effective anti-JEV agent by preventing JEV entry.


Subject(s)
Antiviral Agents/pharmacology , Benzylisoquinolines/pharmacology , Encephalitis Virus, Japanese/drug effects , Encephalitis, Japanese/drug therapy , Lysosomes/metabolism , Receptors, LDL/metabolism , Transient Receptor Potential Channels/drug effects , Animals , Antiviral Agents/therapeutic use , Benzylisoquinolines/therapeutic use , Cell Line , Cell Membrane/metabolism , Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/virology , Exosomes/metabolism , Extracellular Vesicles/metabolism , Humans , Mice , Mice, Inbred BALB C , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Virus Internalization/drug effects
8.
STAR Protoc ; 2(2): 100527, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34027485

ABSTRACT

Small molecular probes designed for photopharmacology and opto-chemogenetics are rapidly gaining widespread recognition for investigations of transient receptor potential canonical (TRPC) channels. This protocol describes the use of three photoswitchable diacylglycerol analogs-PhoDAG-1, PhoDAG-3, and OptoDArG-for ultrarapid activation and deactivation of native TRPC2 channels in mouse vomeronasal sensory neurons and olfactory type B cells, as well as heterologously expressed human TRPC6 channels. Photoconversion can be achieved in mammalian tissue slices and enables all-optical stimulation and shutoff of TRPC channels. For complete details on the use and execution of this protocol, please refer to Leinders-Zufall et al. (2018).


Subject(s)
Cytological Techniques/methods , Diglycerides , Photochemical Processes , Transient Receptor Potential Channels , Animals , Cells, Cultured , Diglycerides/chemistry , Diglycerides/pharmacology , Mice , Olfactory Receptor Neurons/cytology , Transient Receptor Potential Channels/analysis , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism , Vomeronasal Organ/cytology
9.
Molecules ; 26(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804078

ABSTRACT

Sneezing (sternutatio) is a poorly understood polysynaptic physiologic reflex phenomenon. Sneezing has exerted a strange fascination on humans throughout history, and induced sneezing was widely used by physicians for therapeutic purposes, on the assumption that sneezing eliminates noxious factors from the body, mainly from the head. The present contribution examines the various mixtures used for inducing sneezes (remedia sternutatoria) over the centuries. The majority of the constituents of the sneeze-inducing remedies are modulators of transient receptor potential (TRP) channels. The TRP channel superfamily consists of large heterogeneous groups of channels that play numerous physiological roles such as thermosensation, chemosensation, osmosensation and mechanosensation. Sneezing is associated with the activation of the wasabi receptor, (TRPA1), typical ligand is allyl isothiocyanate and the hot chili pepper receptor, (TRPV1), typical agonist is capsaicin, in the vagal sensory nerve terminals, activated by noxious stimulants.


Subject(s)
Sneezing/physiology , Transient Receptor Potential Channels/metabolism , Animals , Capsaicin/pharmacology , Humans , Sneezing/drug effects , Transient Receptor Potential Channels/drug effects
10.
Commun Biol ; 4(1): 293, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674682

ABSTRACT

Monoterpenes are major constituents of plant-derived essential oils and have long been widely used for therapeutic and cosmetic applications. The monoterpenes menthol and camphor are agonists or antagonists for several TRP channels such as TRPM8, TRPV1, TRPV3 and TRPA1. However, which regions within TRPV1 and TRPV3 confer sensitivity to monoterpenes or other synthesized chemicals such as 2-APB are unclear. In this study we identified conserved arginine and glycine residues in the linker between S4 and S5 that are related to the action of these chemicals and validated these findings in molecular dynamics simulations. The involvement of these amino acids differed between TRPV3 and TRPV1 for chemical-induced and heat-evoked activation. These findings provide the basis for characterization of physiological function and biophysical properties of ion channels.


Subject(s)
Membrane Transport Modulators/pharmacology , Monoterpenes/pharmacology , Transient Receptor Potential Channels/drug effects , Amino Acid Sequence , Animals , Arginine , Camphor/chemistry , Camphor/pharmacology , Glycine , HEK293 Cells , Humans , Membrane Potentials , Membrane Transport Modulators/chemistry , Menthol/chemistry , Menthol/pharmacology , Mice , Molecular Dynamics Simulation , Molecular Structure , Monoterpenes/chemistry , Rats , Structure-Activity Relationship , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
11.
Neuroendocrinology ; 111(1-2): 45-69, 2021.
Article in English | MEDLINE | ID: mdl-32028278

ABSTRACT

OBJECTIVE: We examined whether pituitary adenylate cyclase-activating polypeptide (PACAP) excites proopiomelanocortin (POMC) neurons via PAC1 receptor mediation and transient receptor potential cation (TRPC) channel activation. METHODS: Electrophysiological recordings were done in slices from both intact male and ovariectomized (OVX) female PACAP-Cre mice and eGFP-POMC mice. RESULTS: In recordings from POMC neurons in eGFP-POMC mice, PACAP induced a robust inward current and increase in conductance in voltage clamp, and a depolarization and increase in firing in current clamp. These postsynaptic actions were abolished by inhibitors of the PAC1 receptor, TRPC channels, phospholipase C, phosphatidylinositol-3-kinase, and protein kinase C. Estradiol augmented the PACAP-induced inward current, depolarization, and increased firing, which was abrogated by estrogen receptor (ER) antagonists. In optogenetic recordings from POMC neurons in PACAP-Cre mice, high-frequency photostimulation induced inward currents, depolarizations, and increased firing that were significantly enhanced by Gq-coupled membrane ER signaling in an ER antagonist-sensitive manner. Importantly, the PACAP-induced excitation of POMC neurons was notably reduced in obese, high-fat (HFD)-fed males. In vivo experiments revealed that intra-arcuate nucleus (ARC) PACAP as well as chemogenetic and optogenetic stimulation of ventromedial nucleus (VMN) PACAP neurons produced a significant decrease in energy intake accompanied by an increase in energy expenditure, effects blunted by HFD in males and partially potentiated by estradiol in OVX females. CONCLUSIONS: These findings reveal that the PACAP-induced activation of PAC1 receptor and TRPC5 channels at VMN PACAP/ARC POMC synapses is potentiated by estradiol and attenuated under conditions of diet-induced obesity/insulin resistance. As such, they advance our understanding of how PACAP regulates the homeostatic energy balance circuitry under normal and pathophysiological circumstances.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Energy Metabolism/physiology , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Pro-Opiomelanocortin , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Transient Receptor Potential Channels/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Electrophysiological Phenomena , Energy Metabolism/drug effects , Female , Guinea Pigs , Homeostasis , Male , Mice , Mice, Transgenic , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Transient Receptor Potential Channels/drug effects
12.
Fundam Clin Pharmacol ; 35(3): 506-523, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33107619

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting side effect induced by a variety of chemotherapeutic agents. Symptoms are mainly sensory: pain, tingling, numbness, and temperature sensitivity. They may require the tapering of chemotherapy regimens or even their cessation; thus, the prevention/treatment of CIPN is critical to increase effectiveness of cancer treatment. However, CIPN management is mainly based on conventional neuropathic pain treatments, with poor clinical efficacy. Therefore, significant effort is made to identify new pharmacological targets to prevent/treat CIPN. Animal modeling is a key component in predicting human response to drugs and in understanding the pathophysiological mechanisms underlying CIPN. In fact, studies performed in rodents highlighted several pharmacological targets to treat/prevent CIPN. This review provides updated information about ongoing clinical trials testing drugs for the management of CIPN and presents some of their proof-of-concept studies conducted in rodent models. The presented drugs target oxidative stress, renin-angiotensin system, glutamatergic neurotransmission, sphingolipid metabolism, neuronal uptake transporters, nicotinamide adenine dinucleotide metabolism, endocannabinoid system, transient receptor potential channels, and serotoninergic receptors. As some clinical trials focus on the effect of the drugs on pain, others evaluate their efficacy by assessing general neuropathy. Moreover, based on studies conducted in rodent models, it remains unclear if some of the tested drugs act in an antinociceptive fashion or have neuroprotective properties. Thus, further investigations are needed to understand their mechanism of action, as well as a global standardization of the methods used to assess efficacy of new therapeutic strategies in the treatment of CIPN.


Subject(s)
Antineoplastic Agents/adverse effects , Neuralgia/chemically induced , Neuralgia/drug therapy , Animals , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Endocannabinoids/metabolism , Glutamates/drug effects , Humans , NAD/metabolism , Neuralgia/physiopathology , Oxidative Stress/drug effects , Pain/drug therapy , Receptors, Serotonin/drug effects , Renin-Angiotensin System/drug effects , Rodentia , Sphingolipids/metabolism , Transient Receptor Potential Channels/drug effects
13.
Expert Opin Investig Drugs ; 29(11): 1209-1222, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32941080

ABSTRACT

INTRODUCTION: Thermo transient receptor potential (thermoTRP) channels are some of the most intensely pursued therapeutic targets of the past decade. They are considered promising targets of numerous diseases including chronic pain and cancer. Modulators of these proteins, in particular TRPV1-4, TRPM8 and TRPA1, have reached clinical development, but none has been approved for clinical practice yet. AREAS COVERED: The therapeutic potential of targeting thermoTRP channels is discussed. The discussion is centered on our experience and on available data found in SciFinder, PubMed, and ClinicalTrials.gov database from the past decade. This review focuses on the therapeutic progress concerning this family of channels, including strategies to improve their therapeutic index for overcoming adverse effects. EXPERT OPINION: Although thermoTRPs are pivotal drug targets, translation to the clinic has faced two key problems, (i) unforeseen side effects in Phase I trials and, (ii) poor clinical efficacy in Phase II trials. Thus, there is a need for (i) an enhanced understanding of the physiological role of these channels in tissues and organs and (ii) the development of human-based pre-clinical models with higher clinical translation. Furthermore, progress in nanotechnology-based delivery strategies will positively impact thermoTRP human pharmacology.


Subject(s)
Drug Development , Drugs, Investigational/pharmacology , Transient Receptor Potential Channels/drug effects , Animals , Chronic Pain/drug therapy , Chronic Pain/pathology , Drug Delivery Systems , Drugs, Investigational/adverse effects , Humans , Nanotechnology , Neoplasms/drug therapy , Neoplasms/pathology , Transient Receptor Potential Channels/metabolism
14.
J Nat Prod ; 83(9): 2727-2736, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32880179

ABSTRACT

Cannabitwinol (CBDD, 3), the second member of a new class of dimeric phytocannabinoids in which two units are connected by a methylene bridge, was isolated from a hemp (Cannabis sativa L.) industrial extract. The structural characterization of cannabitwinol, complicated by broadening of 1H NMR signals and lack of expected 2D NMR correlations at room temperature, was fully carried out in methanol-d4 at -30 °C. All the attempts to prepare CBDD by reaction of CBD with formaldehyde or its iminium analogue (Eschenmoser salt) failed, suggesting that this sterically congested dimer is the result of enzymatic reactions on the corresponding monomeric acids. Analysis of the cannabitwinol profile of transient receptor potential (TRP) modulation evidenced the impact of dimerization, revealing a selectivity for channels activated by a decrease of temperature (TRPM8 and TRPA1) and the lack of significant affinity for those activated by an increase of temperature (e.g., TRPV1). The putative binding modes of cannabitwinol with TRPA1 and TRPM8 were investigated in detail by a molecular docking study using the homology models of both channels.


Subject(s)
Cannabinoids/chemistry , Cannabinoids/pharmacology , Cannabis/chemistry , Cannabinoids/biosynthesis , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Docking Simulation , Molecular Structure , TRPA1 Cation Channel/drug effects , TRPM Cation Channels/drug effects , TRPV Cation Channels/drug effects , Temperature , Transient Receptor Potential Channels/drug effects
15.
Expert Opin Ther Targets ; 24(11): 1079-1097, 2020 11.
Article in English | MEDLINE | ID: mdl-32972264

ABSTRACT

INTRODUCTION: A myriad of cellular pathophysiological responses are mediated by polymodal ion channels that respond to chemical and physical stimuli such as thermoTRP channels. Intriguingly, these channels are pivotal therapeutic targets with limited clinical pharmacology. In silico methods offer an unprecedented opportunity for discovering new lead compounds targeting thermoTRP channels with improved pharmacological activity and therapeutic index. AREAS COVERED: This article reviews the progress on thermoTRP channel pharmacology because of (i) advances in solving their atomic structure using cryo-electron microscopy and, (ii) progress on computational techniques including homology modeling, molecular docking, virtual screening, molecular dynamics, ADME/Tox and artificial intelligence. Together, they have increased the number of lead compounds with clinical potential to treat a variety of pathologies. We used original and review articles from Pubmed (1997-2020), as well as the clinicaltrials.gov database, containing the terms thermoTRP, artificial intelligence, docking, and molecular dynamics. EXPERT OPINION: The atomic structure of thermoTRP channels along with computational methods constitute a realistic first line strategy for designing drug candidates with improved pharmacology and clinical translation. In silico approaches can also help predict potential side-effects that can limit clinical development of drug candidates. Together, they should provide drug candidates with upgraded therapeutic properties.


Subject(s)
Drug Design , Drug Discovery/methods , Transient Receptor Potential Channels/drug effects , Animals , Artificial Intelligence , Computer Simulation , Cryoelectron Microscopy , Drug Development , Humans , Molecular Docking Simulation , Molecular Targeted Therapy , Transient Receptor Potential Channels/metabolism
16.
Life Sci ; 257: 118112, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32682914

ABSTRACT

AIMS: STW 5 is an herbal drug combination used for the treatment of functional gastrointestinal disorders (FGIDs) with visceral hypersensitivity as the therapy-resistant hallmark. STW 5 has been clinically proven to alleviate visceral hypersensitivity-related symptoms, including abdominal pain, bloating, nausea, and early satiety. However, the molecular and cellular mechanisms underlying the antinociceptive action of STW 5 remain unknown. Here, we investigate the role of STW 5 in the calcium mobilisation of dorsal root ganglion (DRG) sensory neurons. MAIN METHODS: Calcium imaging experiments were performed with freshly dissociated cultured murine DRG neurons isolated from mice by microfluorometry. TRPA1-deficient DRGs, TRPV1-deficient DRGs, TRPA1/V1 double-deficient DRGs, and wild-type DRGs have been used to investigate the role of TRPs ion channels in mediating STW 5 action. KEY FINDINGS: STW 5 (1.74 and 5.8 mg/ml) induced calcium ion influx into DRG neurons in a concentration-dependent manner. Calcium transients were desensitised during repeated exposure to STW 5, an effect that was facilitated in TRPA1-deficient DRGs and less pronounced in TRPV1-deficient DRGs compared to wild-type (WT) DRGs. SIGNIFICANCE: Repeated exposure to STW 5 induced desensitisation of sensory neurons and may ultimately contribute to its proven clinical efficacy against sensory-related symptoms in patients with FGID, including abdominal pain, bloating, nausea, and early satiety. This effect is modulated by the two prominent irritant sensors in nociceptors, TRPA1 and TRPV1.


Subject(s)
Ganglia, Spinal/drug effects , Plant Extracts/pharmacology , Transient Receptor Potential Channels/drug effects , Animals , Calcium/metabolism , Mice , Mice, Inbred C57BL , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
17.
Biomed Res Int ; 2020: 6456805, 2020.
Article in English | MEDLINE | ID: mdl-32337263

ABSTRACT

Despite the wide application of carvacrol (CAR) in medicines, dietary supplements, and foods, there is still insufficient electrophysiological data on the mechanisms of action of CAR, particularly with regard to heart function. Therefore, in this study, we attempted to elucidate whether CAR, whose inhibitory effect on both cardiac and vascular TRPM7 and L-type Ca2+ currents has been demonstrated previously, could modify cardiac electrical activity. We used a combination of optical mapping and microelectrode techniques to track the action potentials (APs) and the spread of electrical activity in a Langendorff-perfused rabbit heart model during atrial/endo/epicardial pacing. Simultaneously, ECG recordings were acquired. Because human trials on CAR are still lacking, we tested the action of CAR on human ventricular preparations obtained from explanted hearts. Activation time (AT), AP duration (APD), and conduction velocity maps were constructed. We demonstrated that at a low concentration (10 µM) of CAR, only marginal changes in the AP parameters were observed. At higher concentrations (≥100 µM), a decrease in AP upstroke velocity (dV/dt max), suggesting inhibition of Na+ current, and APD (at 50 and 90% repolarization) was detected; also slowing in the spread of electrical signals via the atrioventricular node was observed, suggesting impaired functioning of Ca2+ channels. In addition, a decrease in the T-wave amplitude was seen on the ECG, suggesting an impaired repolarization process. Nevertheless, those changes occurred without a significant impact on the resting membrane potential and were reversible. We suggest that CAR might play a role in modulating cardiac electrical activity at high concentrations.


Subject(s)
Cymenes/pharmacology , Heart Ventricles/drug effects , Transient Receptor Potential Channels/drug effects , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Female , Heart Atria/drug effects , Heart Atria/metabolism , Heart Ventricles/metabolism , Humans , Male , Microelectrodes , Middle Aged , Patch-Clamp Techniques/methods , Rabbits , Sodium/metabolism , Transient Receptor Potential Channels/metabolism
18.
Article in English | MEDLINE | ID: mdl-32250774

ABSTRACT

Ion channels underlie electrical excitability in cells and are essential for a variety of functions, most notably neuromuscular and sensory activity. They are also validated targets for a preponderance of approved anthelmintic compounds. Transient receptor potential (TRP) channels constitute an ion channel superfamily whose members play important roles in sensory signaling, regulation of ion homeostasis, organellar trafficking, and other key cellular and organismal activities. Unlike most other ion channels, TRP channels are often polymodal, gated by a variety of mechanisms. Furthermore, TRP channels fall into several classes or subtypes based on sequence and structure. Until recently, there had been very little investigation of the properties and functions of TRP channels from parasitic helminths, including schistosomes, but that situation has changed in the past few years. Indeed, it is now clear that at least some schistosome TRP channels exhibit unusual pharmacological properties, and, intriguingly, both a mammalian and a schistosome TRP channel are activated by praziquantel, the current antischistosomal drug of choice. With the latest release of the Schistosoma mansoni genome database, several changes in predicted TRP channel sequences appeared, some of which were significant. This review updates and reassesses the TRP channel repertoire in S. mansoni, examines recent findings regarding these potential therapeutic targets, and provides guideposts for some of the physiological functions that may be mediated by these channels in schistosomes.


Subject(s)
Schistosoma/physiology , Transient Receptor Potential Channels/physiology , Animals , Anthelmintics/pharmacology , Genes, Helminth , Genome, Helminth , Humans , Ion Channels/drug effects , Ion Channels/genetics , Ion Channels/physiology , Phylogeny , Praziquantel/pharmacology , Schistosoma/drug effects , Schistosoma mansoni/drug effects , Schistosoma mansoni/physiology , Schistosomiasis/drug therapy , TRPA1 Cation Channel/drug effects , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/physiology , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/genetics
19.
ACS Chem Neurosci ; 11(3): 268-290, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31850745

ABSTRACT

Structure-activity relationship studies of a reported menthol-based transient receptor potential cation channel subfamily M member 8 channel (TRPM8) antagonist, guided by computational simulations and structure-based design, uncovers a novel series of TRPM8 antagonists with >10-fold selectivity versus related TRP subtypes. Spiro[4.5]decan-8-yl analogue 14 inhibits icilin-evoked Ca2+ entry in HEK-293 cells stably expressing human TRPM8 (hTRPM8) with an IC50 of 2.4 ± 1.0 nM, while in whole-cell patch-clamp recordings this analogue inhibits menthol-evoked currents with a hTRPM8 IC50 of 64 ± 2 nM. Molecular dynamics (MD) simulations of compound 14 in our homology model of hTRPM8 suggest that this antagonist forms extensive hydrophobic contacts within the orthosteric site. In the wet dog shakes (WDS) assay, compound 14 dose-dependently blocks icilin-triggered shaking behaviors in mice. Upon local administration, compound 14 dose dependently inhibits cold allodynia evoked by the chemotherapy oxaliplatin in a murine model of peripheral neuropathy at microgram doses. Our findings suggest that 14 and other biphenyl amide analogues within our series can find utility as potent antagonist chemical probes derived from (-)-menthol as well as small molecule therapeutic scaffolds for chemotherapy-induced peripheral neuropathy (CIPN) and other sensory neuropathies.


Subject(s)
Biphenyl Compounds/antagonists & inhibitors , Hyperalgesia/drug therapy , Peripheral Nervous System Diseases/drug therapy , Structure-Activity Relationship , TRPM Cation Channels/metabolism , Amides , Calcium/metabolism , HEK293 Cells , Humans , Menthol/analogs & derivatives , Patch-Clamp Techniques/methods , TRPM Cation Channels/drug effects , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism
20.
Behav Pharmacol ; 31(5): 413-434, 2020 08.
Article in English | MEDLINE | ID: mdl-31833970

ABSTRACT

The transient receptor potential (TRP) channel superfamily is comprised of a large group of cation-permeable channels, which display an extraordinary diversity of roles in sensory signaling and are involved in plethora of animal behaviors. These channels are activated through a wide variety of mechanisms and participate in virtually every sensory modality. Modulating TRP channel activity provides an important way to regulate membrane excitability and intracellular calcium levels. This is reflected by the fact that small molecule compounds modulating different TRPs have all entered clinical trials for a variety of diseases. The role of TRPs will be further elucidated in complex diseases of the nervous, intestinal, renal, urogenital, respiratory, and cardiovascular systems in diverse therapeutic areas including pain and itch, headache, pulmonary function, oncology, neurology, visceral organs, and genetic diseases. This review focuses on recent developments in the TRP ion channel-related area and highlights evidence supporting TRP channels as promising targets for new analgesic drugs for therapeutic intervention. This review presents a variety of: (1) phylogeny aspects of TRP channels; (2) some structural and functional characteristics of TRPs; (3) a general view and short characteristics of main seven subfamilies of TRP channels; (4) the evidence for consider TRP channels as therapeutic and analgesic targets; and finally (5) further perspectives of TRP channels research.


Subject(s)
Analgesics/pharmacology , Behavior, Animal/physiology , Molecular Targeted Therapy/methods , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/physiology , Analgesics/therapeutic use , Animals , Humans , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...