Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Biomedicines ; 11(3)2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36979754

ABSTRACT

Neuropathic pain is a debilitating condition affecting around 8% of the adult population in the UK. The pathophysiology is complex and involves a wide range of processes, including alteration of neuronal excitability and synaptic transmission, dysregulated intracellular signalling and activation of pro-inflammatory immune and glial cells. In the past 15 years, multiple miRNAs-small non-coding RNA-have emerged as regulators of neuropathic pain development. They act by binding to target mRNAs and preventing the translation into proteins. Due to their short sequence (around 22 nucleotides in length), they can have hundreds of targets and regulate several pathways. Several studies on animal models have highlighted numerous miRNAs that play a role in neuropathic pain development at various stages of the nociceptive pathways, including neuronal excitability, synaptic transmission, intracellular signalling and communication with non-neuronal cells. Studies on animal models do not always translate in the clinic; fewer studies on miRNAs have been performed involving human subjects with neuropathic pain, with differing results depending on the specific aetiology underlying neuropathic pain. Further studies using human tissue and liquid samples (serum, plasma, saliva) will help highlight miRNAs that are relevant to neuropathic pain diagnosis or treatment, as biomarkers or potential drug targets.

2.
J Pharmacol Exp Ther ; 383(2): 117-128, 2022 11.
Article in English | MEDLINE | ID: mdl-36116796

ABSTRACT

Using synaptosomes purified from the brains of two transgenic mouse models overexpressing mutated human tau (TgP301S and Tg4510) and brains of patients with sporadic Alzheimer's disease, we showed that aggregated and hyperphosphorylated tau was both present in purified synaptosomes and released in a calcium- and synaptosome-associated protein of 25 kDa (SNAP25)-dependent manner. In all mouse and human synaptosomal preparations, tau release was inhibited by the selective metabotropic glutamate receptor 2/3 (mGluR2/3) agonist LY379268, an effect prevented by the selective mGlu2/3 antagonist LY341495. LY379268 was also able to block pathologic tau propagation between primary neurons in an in vitro microfluidic cellular model. These novel results are transformational for our understanding of the molecular mechanisms mediating tau release and propagation at synaptic terminals in Alzheimer's disease and suggest that these processes could be inhibited therapeutically by the selective activation of presynaptic G protein-coupled receptors. SIGNIFICANCE STATEMENT: Pathological tau release and propagation are key neuropathological events underlying cognitive decline in Alzheimer's disease patients. This paper describes the role of regulated exocytosis, and the soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) protein SNAP25, in mediating tau release from rodent and human synaptosomes. This paper also shows that a selective mGluR2/3 agonist is highly effective in blocking tau release from synaptosomes and tau propagation between neurons, opening the way to the discovery of novel therapeutic approaches to this devastating disease.


Subject(s)
Alzheimer Disease , Receptors, Metabotropic Glutamate , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Animals , Calcium/metabolism , Exocytosis , Humans , Mice , N-Ethylmaleimide-Sensitive Proteins/metabolism , N-Ethylmaleimide-Sensitive Proteins/pharmacology , Receptors, Metabotropic Glutamate/metabolism , SNARE Proteins/metabolism , SNARE Proteins/pharmacology , Synaptosomes/metabolism
3.
Pain Med ; 23(6): 1084-1094, 2022 05 30.
Article in English | MEDLINE | ID: mdl-34850195

ABSTRACT

BACKGROUND: Fibromyalgia syndrome (FMS) is the most common chronic widespread pain condition in rheumatology. Until recently, no clear pathophysiological mechanism for fibromyalgia had been established, resulting in management challenges. Recent research has indicated that serum immunoglobulin Gs (IgGs) may play a role in FMS. We undertook a research prioritisation exercise to identify the most pertinent research approaches that may lead to clinically implementable outputs. METHODS: Research priority setting was conducted in five phases: situation analysis; design; expert group consultation; interim recommendations; consultation and revision. A dialogue model was used, and an international multi-stakeholder expert group was invited. Clinical, patient, industry, funder, and scientific expertise was represented throughout. Recommendation-consensus was determined via a voluntary closed eSurvey. Reporting guideline for priority setting of health research were employed to support implementation and maximise impact. RESULTS: Arising from the expert group consultation (n = 29 participants), 39 interim recommendations were defined. A response rate of 81.5% was achieved in the consensus survey. Six recommendations were identified as high priority- and 15 as medium level priority. The recommendations range from aspects of fibromyalgia features that should be considered in future autoantibody research, to specific immunological investigations, suggestions for trial design in FMS, and therapeutic interventions that should be assessed in trials. CONCLUSIONS: By applying the principles of strategic priority setting we directed research towards that which is implementable, thereby expediating the benefit to the FMS patient population. These recommendations are intended for patients, international professionals and grant-giving bodies concerned with research into causes and management of patients with fibromyalgia syndrome.


Subject(s)
Chronic Pain , Fibromyalgia , Autoantibodies , Fibromyalgia/therapy , Humans , Immunoglobulin G , Surveys and Questionnaires
4.
Pain ; 161(9): 2155-2166, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32379221

ABSTRACT

ABSTRACT: Rheumatoid arthritis-associated pain is poorly managed, often persisting when joint inflammation is pharmacologically controlled. Comparably, in the mouse K/BxN serum-transfer model of inflammatory arthritis, hind paw nociceptive hypersensitivity occurs with ankle joint swelling (5 days after immunisation) persisting after swelling has resolved (25 days after immunisation). In this study, lipid mediator (LM) profiling of lumbar dorsal root ganglia (DRG), the site of sensory neuron cell bodies innervating the ankle joints, 5 days and 25 days after serum transfer demonstrated a shift in specialised proresolving LM profiles. Persistent nociception without joint swelling was associated with low concentrations of the specialised proresolving LM Maresin 1 (MaR1) and high macrophage numbers in DRG. MaR1 application to cultured DRG neurons inhibited both capsaicin-induced increase of intracellular calcium ions and release of calcitonin gene-related peptide in a dose-dependent manner. Furthermore, in peritoneal macrophages challenged with lipopolysaccharide, MaR1 reduced proinflammatory cytokine expression. Systemic MaR1 administration caused sustained reversal of nociceptive hypersensitivity and reduced inflammatory macrophage numbers in DRG. Unlike gabapentin, which was used as positive control, systemic MaR1 did not display acute antihyperalgesic action. Therefore, these data suggest that MaR1 effects observed after K/BxN serum transfer relate to modulation of macrophage recruitment, more likely than to direct actions on sensory neurons. Our study highlights that, in DRG, aberrant proresolution mechanisms play a key role in arthritis joint pain dissociated from joint swelling, opening novel approaches for rheumatoid arthritis pain treatment.


Subject(s)
Ganglia, Spinal , Hyperalgesia , Animals , Calcitonin Gene-Related Peptide , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Macrophages , Mice , Pain
5.
Assay Drug Dev Technol ; 17(7): 310-321, 2019 10.
Article in English | MEDLINE | ID: mdl-31634018

ABSTRACT

Relief from chronic pain continues to represent a large unmet need. The voltage-gated potassium channel Kv7.2/7.3, also known as KCNQ2/3, is a key contributor to the control of resting membrane potential and excitability in nociceptive neurons and represents a promising target for potential therapeutics. In this study, we present a medium throughput electrophysiological assay for the identification and characterization of modulators of Kv7.2/7.3 channels, using the IonWorks Barracuda™ automated voltage clamp platform. The assay combines a family of voltage steps used to construct conductance curves with a unique analysis method. Kv7.2/7.3 modulators shift the activation voltage and/or change the maximal conductance of the current, and both parameters have been used to quantify compound mediated effects. Both effects are expected to modulate neuronal excitability in vivo. The analysis method described assigns a single potency value that combines changes in activation voltage and maximal conductance and is expected to predict compound mediated changes in excitability.


Subject(s)
Aminopyridines/analysis , Carbamates/analysis , Drug Development , High-Throughput Screening Assays/instrumentation , Patch-Clamp Techniques/instrumentation , Phenylenediamines/analysis , Aminopyridines/pharmacology , Carbamates/pharmacology , Cells, Cultured , Electrophysiological Phenomena , HEK293 Cells , Humans , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Phenylenediamines/pharmacology
7.
J Pharmacol Exp Ther ; 369(3): 345-363, 2019 06.
Article in English | MEDLINE | ID: mdl-30910921

ABSTRACT

Nonselective glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonists are efficacious in chronic pain but have significant tolerability issues, likely arising from the ubiquitous expression of AMPA receptors in the central nervous system (CNS). Recently, LY3130481 has been shown to selectively block AMPA receptors coassembled with the auxiliary protein, transmembrane AMPA receptor regulatory protein (TARP) γ8, which is highly expressed in the hippocampus but also in pain pathways, including anterior cingulate (ACC) and somatosensory cortices and the spinal cord, suggesting that selective blockade of γ8/AMPA receptors may suppress nociceptive signaling with fewer CNS side effects. The potency of LY3130481 on recombinant γ8-containing AMPA receptors was modulated by coexpression with other TARPs; γ2 subunits affected activity more than γ3 subunits. Consistent with these findings, LY3130481 had decreasing potency on receptors from rat hippocampal, cortical, spinal cord, and cerebellar neurons that was replicated in tissue from human brain. LY3130481 partially suppressed, whereas the nonselective AMPA antagonist GYKI53784 completely blocked, AMPA receptor-dependent excitatory postsynaptic potentials in ACC and spinal neurons in vitro. Similarly, LY3130481 attenuated short-term synaptic plasticity in spinal sensory neurons in vivo in response to stimulation of peripheral afferents. LY3130481 also significantly reduced nocifensive behaviors after intraplantar formalin that was correlated with occupancy of CNS γ8-containing AMPA receptors. In addition, LY3130481 dose-dependently attenuated established gait impairment after joint damage and tactile allodynia after spinal nerve ligation, all in the absence of motor side effects. Collectively, these data demonstrate that LY3130481 can suppress excitatory synaptic transmission and plasticity in pain pathways containing γ8/AMPA receptors and significantly reduce nocifensive behaviors, suggesting a novel, effective, and safer therapy for chronic pain conditions.


Subject(s)
Calcium Channels/metabolism , Chronic Pain/drug therapy , Chronic Pain/metabolism , Molecular Targeted Therapy , Receptors, AMPA/metabolism , Animals , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Chronic Pain/physiopathology , Male , Neuronal Plasticity/drug effects , Nociception/drug effects , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Tissue Distribution
8.
Br J Pharmacol ; 176(1): 110-126, 2019 01.
Article in English | MEDLINE | ID: mdl-30276808

ABSTRACT

BACKGROUND AND PURPOSE: We aimed to identify and develop novel, selective muscarinic M1 receptor agonists as potential therapeutic agents for the symptomatic treatment of Alzheimer's disease. EXPERIMENTAL APPROACH: We developed and utilized a novel M1 receptor occupancy assay to drive a structure activity relationship in a relevant brain region while simultaneously tracking drug levels in plasma and brain to optimize for central penetration. Functional activity was tracked in relevant native in vitro assays allowing translational (rat-human) benchmarking of structure-activity relationship molecules to clinical comparators. KEY RESULTS: Using this paradigm, we identified a series of M1 receptor selective molecules displaying desirable in vitro and in vivo properties and optimized key features, such as central penetration while maintaining selectivity and a partial agonist profile. From these compounds, we selected spiropiperidine 1 (SPP1). In vitro, SPP1 is a potent, partial agonist of cortical and hippocampal M1 receptors with activity conserved across species. SPP1 displays high functional selectivity for M1 receptors over native M2 and M3 receptor anti-targets and over a panel of other targets. Assessment of central target engagement by receptor occupancy reveals SPP1 significantly and dose-dependently occupies rodent cortical M1 receptors. CONCLUSIONS AND IMPLICATIONS: We report the discovery of SPP1, a novel, functionally selective, brain penetrant partial orthosteric agonist at M1 receptors, identified by a novel receptor occupancy assay. SPP1 is amenable to in vitro and in vivo study and provides a valuable research tool to further probe the role of M1 receptors in physiology and disease.


Subject(s)
Osteopontin/agonists , Piperidines/pharmacology , Receptor, Muscarinic M1/agonists , Spiro Compounds/pharmacology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Piperidines/chemistry , Rats , Rats, Sprague-Dawley , Spiro Compounds/chemistry , Structure-Activity Relationship , Xenopus
9.
Drug Discov Today ; 24(2): 533-543, 2019 02.
Article in English | MEDLINE | ID: mdl-30395928

ABSTRACT

It is important in drug discovery to demonstrate that activity of novel drugs found by screening on recombinant receptors translates to activity on native human receptors in brain areas affected by disease. In this review, we summarise the development and use of the microtransplantation technique. Native receptors are reconstituted from human brain tissues into oocytes from the frog Xenopus laevis where they can be functionally assessed. Oocytes microtransplanted with hippocampal tissue from an epileptic patient were used to demonstrate that new antiepileptic agents act on receptors in diseased tissue. Furthermore, frozen post-mortem human tissues were used to show that drugs are active on receptors in brain areas associated with a disease; but not in areas associated with side effects.


Subject(s)
Brain/metabolism , Oocytes/metabolism , Receptors, Cell Surface/physiology , Transplantation, Heterologous/methods , Animals , Drug Discovery , Humans
10.
Biochem Biophys Res Commun ; 495(1): 481-486, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29127015

ABSTRACT

Muscarinic acetylcholine M1 receptors play an important role in synaptic plasticity in the hippocampus and cortex. Potentiation of NMDA receptors as a consequence of muscarinic acetylcholine M1 receptor activation is a crucial event mediating the cholinergic modulation of synaptic plasticity, which is a cellular mechanism for learning and memory. In Alzheimer's disease, the cholinergic input to the hippocampus and cortex is severely degenerated, and agonists or positive allosteric modulators of M1 receptors are therefore thought to be of potential use to treat the deficits in cognitive functions in Alzheimer's disease. In this study we developed a simple system in which muscarinic modulation of NMDA receptors can be studied in vitro. Human M1 receptors and NR1/2B NMDA receptors were co-expressed in Xenopus oocytes and various muscarinic agonists were assessed for their modulatory effects on NMDA receptor-mediated responses. As expected, NMDA receptor-mediated responses were potentiated by oxotremorine-M, oxotremorine or xanomeline when the drugs were applied between subsequent NMDA responses, an effect which was fully blocked by the muscarinic receptor antagonist atropine. However, in oocytes expressing NR1/2B NMDA receptors but not muscarinic M1 receptors, oxotremorine-M co-applied with NMDA also resulted in a potentiation of NMDA currents and this effect was not blocked by atropine, demonstrating that oxotremorine-M is able to directly potentiate NMDA receptors. Oxotremorine, which is a close analogue of oxotremorine-M, and xanomeline, a chemically distinct muscarinic agonist, did not potentiate NMDA receptors by this direct mechanism. Comparing the chemical structures of the three different muscarinic agonists used in this study suggests that the tri-methyl ammonium moiety present in oxotremorine-M is important for the compound's interaction with NMDA receptors.


Subject(s)
Muscarinic Agonists/pharmacology , Oxotremorine/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/agonists , Animals , Humans , Muscarinic Agonists/chemistry , Oxotremorine/chemistry , Oxotremorine/pharmacology , Phosphorylation/drug effects , Protein Kinase C/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Xenopus
11.
Nat Neurosci ; 21(1): 130-138, 2018 01.
Article in English | MEDLINE | ID: mdl-29203896

ABSTRACT

The postsynaptic proteome of excitatory synapses comprises ~1,000 highly conserved proteins that control the behavioral repertoire, and mutations disrupting their function cause >130 brain diseases. Here, we document the composition of postsynaptic proteomes in human neocortical regions and integrate it with genetic, functional and structural magnetic resonance imaging, positron emission tomography imaging, and behavioral data. Neocortical regions show signatures of expression of individual proteins, protein complexes, biochemical and metabolic pathways. We characterized the compositional signatures in brain regions involved with language, emotion and memory functions. Integrating large-scale GWAS with regional proteome data identifies the same cortical region for smoking behavior as found with fMRI data. The neocortical postsynaptic proteome data resource can be used to link genetics to brain imaging and behavior, and to study the role of postsynaptic proteins in localization of brain functions.


Subject(s)
Neocortex/pathology , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Synaptosomes/metabolism , Animals , Computational Biology , Female , Humans , Image Processing, Computer-Assisted , Male , Membrane Potentials/genetics , Microinjections , Neocortex/diagnostic imaging , Nerve Tissue Proteins/genetics , Oocytes , Oxygen/blood , Patch-Clamp Techniques , Positron-Emission Tomography , Proteomics , Stroke/pathology , Synapses/ultrastructure , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
12.
Mol Pain ; 13: 1744806917745179, 2017.
Article in English | MEDLINE | ID: mdl-29166836

ABSTRACT

Background The Nav1.7 subtype of voltage-gated sodium channels is specifically expressed in sensory and sympathetic ganglia neurons where it plays an important role in the generation and transmission of information related to pain sensation. Human loss or gain-of-function mutations in the gene encoding Nav1.7 channels (SCN9A) are associated with either absence of pain, as reported for congenital insensitivity to pain, or with exacerbation of pain, as reported for primary erythromelalgia and paroxysmal extreme pain disorder. Based on this important human genetic evidence, numerous drug discovery efforts are ongoing in search for Nav1.7 blockers as a novel therapeutic strategy to treat pain conditions. Results We are reporting here a novel approach to study Nav1.7 function in cultured rat sensory neurons. We used live cell imaging combined with electrical field stimulation to evoke and record action potential-driven calcium transients in the neurons. We have shown that the tarantula venom peptide Protoxin-II, a known Nav1.7 subtype selective blocker, inhibited electrical field stimulation-evoked calcium responses in dorsal root ganglia neurons with an IC50 of 72 nM, while it had no activity in embryonic hippocampal neurons. The results obtained in the live cell imaging assay were supported by patch-clamp studies as well as by quantitative PCR and Western blotting experiments that confirmed the presence of Nav1.7 mRNA and protein in dorsal root ganglia but not in embryonic hippocampal neurons. Conclusions The findings presented here point to a selective effect of Protoxin-II in sensory neurons and helped to validate a new method for investigating and comparing Nav1.7 pharmacology in sensory versus central nervous system neurons. This will help in the characterisation of the selectivity of novel Nav1.7 modulators using native ion channels and will provide the basis for the development of higher throughput models for enabling pain-relevant phenotypic screening.


Subject(s)
Electric Stimulation/methods , Ganglia, Spinal/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sensory Receptor Cells/metabolism , Animals , Calcium/metabolism , Ganglia, Spinal/drug effects , Hippocampus/metabolism , Male , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/pharmacology
13.
Drug Deliv Transl Res ; 7(1): 11-15, 2017 02.
Article in English | MEDLINE | ID: mdl-27553192

ABSTRACT

Intranasally administered regular insulin and insulin aspart have shown cognitive benefit for patients with Alzheimer's disease (AD). To support development of intranasally administered insulin analogs for AD, the central disposition of intranasal insulin lispro in the cerebrospinal fluid (CSF) of healthy volunteers was investigated. Healthy volunteers (N = 8) received two sequential doses of intranasal insulin lispro (48 or 80 IU followed by 160 IU) by Aero Pump in an open-label, single-period study with serial CSF and serum sampling over 5 hours after each dose. CSF insulin lispro was also measured in beagle dogs (N = 6/dose group) that received either 24 IU/kg (equivalent local nasal (IU/cm2) dose to the human 160 IU dose) or 192 IU/kg intranasally, using the same device. Insulin lispro was measured in the CSF and serum using a validated enzyme-linked immunosorbent assay method, and pharmacokinetic parameters were calculated by standard noncompartmental methods. Intranasal administration of insulin lispro was well tolerated. Insulin lispro concentrations in the CSF of humans at all dose levels were below the limit of quantification. Serum insulin lispro concentrations were quantifiable only up to 1-2 hours in the majority of subjects. In contrast to insulin lispro in the CSF of humans, insulin lispro was detectable in the CSF at both dose levels in dogs, and serum concentrations of insulin lispro were generally higher in dogs than in healthy volunteers. The absence of insulin lispro in CSF from healthy volunteers and the lack of robust exposure-response analyses will hinder the development of intranasally administered insulin lispro for AD.


Subject(s)
Hypoglycemic Agents/cerebrospinal fluid , Insulin Lispro/cerebrospinal fluid , Administration, Intranasal , Animals , Dogs , Healthy Volunteers , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Insulin Lispro/administration & dosage , Insulin Lispro/blood , Insulin Lispro/pharmacokinetics , Male , Middle Aged
14.
Nat Med ; 22(12): 1496-1501, 2016 12.
Article in English | MEDLINE | ID: mdl-27820603

ABSTRACT

Pharmacological manipulation of specific neural circuits to optimize therapeutic index is an unrealized goal in neurology and psychiatry. AMPA receptors are important for excitatory synaptic transmission, and their antagonists are antiepileptic. Although efficacious, AMPA-receptor antagonists, including perampanel (Fycompa), the only approved antagonist for epilepsy, induce dizziness and motor impairment. We hypothesized that blockade of forebrain AMPA receptors without blocking cerebellar AMPA receptors would be antiepileptic and devoid of motor impairment. Taking advantage of an AMPA receptor auxiliary protein, TARP γ-8, which is selectively expressed in the forebrain and modulates the pharmacological properties of AMPA receptors, we discovered that LY3130481 selectively antagonized recombinant and native AMPA receptors containing γ-8, but not γ-2 (cerebellum) or other TARP members. Two amino acid residues unique to γ-8 determined this selectivity. We also observed antagonism of AMPA receptors expressed in hippocampal, but not cerebellar, tissue from an patient with epilepsy. Corresponding to this selective activity, LY3130481 prevented multiple seizure types in rats and mice and without motor side effects. These findings demonstrate the first rationally discovered molecule targeting specific neural circuitries for therapeutic advantage.


Subject(s)
Anticonvulsants/pharmacology , Benzothiazoles/pharmacology , Cerebellum/drug effects , Epilepsy/drug therapy , Prosencephalon/drug effects , Pyrazoles/pharmacology , Pyridones/pharmacology , Receptors, AMPA/antagonists & inhibitors , Animals , Anticonvulsants/adverse effects , Calcium Channels/metabolism , Cerebellum/metabolism , Convulsants/toxicity , Disease Models, Animal , Dizziness/chemically induced , Epilepsy/chemically induced , Mice , Nitriles , Pentylenetetrazole/toxicity , Prosencephalon/metabolism , Pyridones/adverse effects , Rats , Receptors, AMPA/metabolism , Seizures/chemically induced , Seizures/drug therapy
15.
Eur J Pharmacol ; 791: 221-228, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27590358

ABSTRACT

Inhibition of KCNQ (Kv7) potassium channels by activation of muscarinic acetylcholine receptors has been well established, and the ion currents through these channels have been long known as M-currents. We found that this cross-talk can be reconstituted in Xenopus oocytes by co-transfection of human recombinant muscarinic M1 receptors and KCNQ2/3 potassium channels. Application of the muscarinic acetylcholine receptor agonist Oxotremorine-methiodide (Oxo-M) between voltage pulses to activate KCNQ2/3 channels caused inhibition of the subsequent KCNQ2/3 responses. This effect of Oxo-M was blocked by the muscarinic acetylcholine receptor antagonist atropine. We also found that KCNQ2/3 currents were inhibited when Oxo-M was applied during an ongoing KCNQ2/3 response, an effect that was not blocked by atropine, suggesting that Oxo-M inhibits KCNQ2/3 channels directly. Indeed, also in oocytes that were transfected with only KCNQ2/3 channels, but not with muscarinic M1 receptors, Oxo-M inhibited the KCNQ2/3 response. These results show that besides the usual muscarinic acetylcholine receptor-mediated inhibition, Oxo-M also inhibits KCNQ2/3 channels by a direct mechanism. We subsequently tested xanomeline, which is a chemically distinct muscarinic acetylcholine receptor agonist, and oxotremorine, which is a close analogue of Oxo-M. Both compounds inhibited KCNQ2/3 currents via activation of M1 muscarinic acetylcholine receptors but, in contrast to Oxo-M, they did not directly inhibit KCNQ2/3 channels. Xanomeline and oxotremorine do not contain a positively charged trimethylammonium moiety that is present in Oxo-M, suggesting that such a charged moiety could be a crucial component mediating this newly described direct inhibition of KCNQ2/3 channels.


Subject(s)
KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ3 Potassium Channel/antagonists & inhibitors , Oxotremorine/analogs & derivatives , Potassium Channel Blockers/pharmacology , Animals , Humans , Oxotremorine/pharmacology , Pyridines/pharmacology , Receptor, Muscarinic M1/metabolism , Thiadiazoles/pharmacology , Xenopus
16.
Physiol Rep ; 4(14)2016 07.
Article in English | MEDLINE | ID: mdl-27462070

ABSTRACT

Connexin (Cx) proteins and gap junctions support the formation of neuronal and glial syncytia that are linked to different forms of rhythmic firing and oscillatory activity in the CNS. In this study, quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to profile developmental expression of two specific Cx proteins, namely glial Cx43 and neuronal Cx36, in postnatal lumbar spinal cord aged 4, 7, and 14 days. Extracellular electrophysiology was used to determine the contribution of Cx36 and Cx43 to a previously described form of 4-aminopyridine (4-AP)-induced 4-12 Hz rhythmic activity within substantia gelatinosa (SG) of rat neonatal dorsal horn (DH) in vitro. The involvement of Cx36 and Cx43 was probed pharmacologically using quinine, a specific uncoupler of Cx36 and the mimetic peptide blocker Gap 26 which targets Cx43. After establishment of 4-12 Hz rhythmic activity by 4-AP (25 µmol/L), coapplication of quinine (250 µmol/L) reduced 4-AP-induced 4-12 Hz rhythmic activity (P < 0.05). Preincubation of spinal cord slices with Gap 26 (100 µmol/L), compromised the level of 4-AP-induced 4-12 Hz rhythmic activity in comparison with control slices preincubated in ACSF alone (P < 0.05). Conversely, the nonselective gap junction "opener" trimethylamine (TMA) enhanced 4-12 Hz rhythmic behavior (P < 0.05), further supporting a role for Cx proteins and gap junctions. These data have defined a physiological role for Cx36 and Cx43 in rhythmic firing in SG, a key nociceptive processing area of DH. The significance of these data in the context of pain and Cx proteins as a future analgesic drug target requires further study.


Subject(s)
Connexin 43/physiology , Connexins/physiology , Spinal Cord Dorsal Horn/metabolism , 4-Aminopyridine/pharmacology , Animals , Connexin 43/genetics , Connexins/genetics , Gap Junctions/drug effects , Gap Junctions/physiology , Gene Expression Regulation, Developmental/physiology , Lumbar Vertebrae , Male , Potassium Channel Blockers/pharmacology , Rats, Wistar , Spinal Cord Dorsal Horn/drug effects , Substantia Gelatinosa/drug effects , Substantia Gelatinosa/metabolism , Tissue Culture Techniques , Gap Junction delta-2 Protein
17.
J Neurochem ; 138(3): 384-96, 2016 08.
Article in English | MEDLINE | ID: mdl-27216696

ABSTRACT

Disruption in the expression and function of synaptic proteins, and ion channels in particular, is critical in the pathophysiology of human neuropsychiatric and neurodegenerative diseases. However, very little is known regarding the functional and pharmacological properties of native synaptic human ion channels, and their potential changes in pathological conditions. Recently, an electrophysiological technique has been enabled for studying the functional and pharmacological properties of ion channels present in crude membrane preparation obtained from post-mortem frozen brains. We here extend these studies by showing that human synaptic ion channels also can be studied in this way. Synaptosomes purified from different regions of rodent and human brain (control and Alzheimer's) were characterized biochemically for enrichment of synaptic proteins, and expression of ion channel subunits. The same synaptosomes were also reconstituted in Xenopus oocytes, in which the functional and pharmacological properties of the native synaptic ion channels were characterized using the voltage clamp technique. We show that we can detect GABA, (RS)-α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and NMDA receptors, and modulate them pharmacologically with selective agonists, antagonists, and allosteric modulators. Furthermore, changes in ion channel expression and function were detected in synaptic membranes from Alzheimer's brains. Our present results demonstrate the possibility to investigate synaptic ion channels from healthy and pathological brains. This method of synaptosomes preparation and injection into oocytes is a significant improvement over the earlier method. It opens the way to directly testing, on native ion channels, the effects of novel drugs aimed at modulating important classes of synaptic targets. Disruption in the expression and function of synaptic ion channels is critical in the pathophysiology of human neurodegenerative diseases. We here show that synaptosomes purified from rodent and human frozen brain (control and Alzheimer disease) can be studied both biochemically and functionally. This method opens the way to directly testing the effects of novel drugs on native ion channels.


Subject(s)
Brain/metabolism , Ion Channels/metabolism , Oocytes/metabolism , Synaptosomes/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Female , Humans , Patch-Clamp Techniques/methods , Rats, Wistar , Receptors, GABA-A/metabolism , Xenopus laevis , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
18.
J Pharmacol Exp Ther ; 355(2): 247-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26324167

ABSTRACT

The citric acid cycle intermediate citrate plays a crucial role in metabolic processes such as fatty acid synthesis, glucose metabolism, and ß-oxidation. Citrate is imported from the circulation across the plasma membrane into liver cells mainly by the sodium-dependent citrate transporter (NaCT; SLC13A5). Deletion of NaCT from mice led to metabolic changes similar to caloric restriction; therefore, NaCT has been proposed as an attractive therapeutic target for the treatment of obesity and type 2 diabetes. In this study, we expressed mouse and human NaCT into Xenopus oocytes and examined some basic functional properties of those transporters. Interestingly, striking differences were found between mouse and human NaCT with respect to their sensitivities to citric acid cycle intermediates as substrates for these transporters. Mouse NaCT had at least 20- to 800-fold higher affinity for these intermediates than human NaCT. Mouse NaCT is fully active at physiologic plasma levels of citrate, but its human counterpart is not. Replacement of extracellular sodium by other monovalent cations revealed that human NaCT was markedly less dependent on extracellular sodium than mouse NaCT. The low sensitivity of human NaCT for citrate raises questions about the translatability of this target from the mouse to the human situation and raises doubts about the validity of this transporter as a therapeutic target for the treatment of metabolic diseases in humans.


Subject(s)
Citric Acid Cycle , Dicarboxylic Acid Transporters/physiology , Symporters/physiology , Animals , Cations, Monovalent , Choline/metabolism , Dicarboxylic Acid Transporters/genetics , Female , Humans , Lithium/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Sodium/metabolism , Substrate Specificity , Symporters/genetics , Xenopus laevis
19.
PLoS One ; 10(6): e0130572, 2015.
Article in English | MEDLINE | ID: mdl-26086615

ABSTRACT

The existence of α7ß2 nicotinic acetylcholine receptors (nAChRs) has recently been demonstrated in both the rodent and human brain. Since α7-containing nAChRs are promising drug targets for schizophrenia and Alzheimer's disease, it is critical to determine whether α7ß2 nAChRs are present in the human brain, in which brain areas, and whether they differ functionally from α7 nAChR homomers. We used α-bungarotoxin to affinity purify α7-containing nAChRs from surgically excised human temporal cortex, and found that α7 subunits co-purify with ß2 subunits, indicating the presence of α7ß2 nAChRs in the human brain. We validated these results by demonstrating co-purification of ß2 from wild-type, but not α7 or ß2 knock-out mice. The pharmacology and kinetics of human α7ß2 nAChRs differed significantly from that of α7 homomers in response to nAChR agonists when expressed in Xenopus oocytes and HEK293 cells. Notably, α7ß2 heteromers expressed in HEK293 cells display markedly slower rise and decay phases. These results demonstrate that α7 subunits in the human brain form heteromeric complexes with ß2 subunits, and that human α7ß2 nAChR heteromers respond to nAChR agonists with a unique pharmacology and kinetic profile. α7ß2 nAChRs thus represent an alternative mechanism for the reported clinical efficacy of α7 nAChR ligands.


Subject(s)
Cerebral Cortex/metabolism , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cerebral Cortex/pathology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/genetics , Xenopus laevis/growth & development , Xenopus laevis/metabolism , alpha7 Nicotinic Acetylcholine Receptor/deficiency , alpha7 Nicotinic Acetylcholine Receptor/genetics
20.
PLoS One ; 10(4): e0125116, 2015.
Article in English | MEDLINE | ID: mdl-25906356

ABSTRACT

Neurons derived from human induced pluripotent stem cells (iPSCs) represent a potentially valuable tool for the characterisation of neuronal receptors and ion channels. Previous studies on iPSC-derived neuronal cells have reported the functional characterisation of a variety of receptors and ion channels, including glutamate receptors, γ-aminobutyric acid (GABA) receptors and several voltage-gated ion channels. In the present study we have examined the expression and functional properties of nicotinic acetylcholine receptors (nAChRs) in human iPSC-derived neurons. Gene expression analysis indicated the presence of transcripts encoding several nAChR subunits, with highest levels detected for α3-α7, ß1, ß2 and ß4 subunits (encoded by CHRNA3-CHRNA7, CHRNB1, CHRNB2 and CHRNB4 genes). In addition, similarly high transcript levels were detected for the truncated dupα7 subunit transcript, encoded by the partially duplicated gene CHRFAM7A, which has been associated with psychiatric disorders such as schizophrenia. The functional properties of these nAChRs have been examined by calcium fluorescence and by patch-clamp recordings. The data obtained suggest that the majority of functional nAChRs expressed in these cells have pharmacological properties typical of α7 receptors. Large responses were induced by a selective α7 agonist (compound B), in the presence of the α7-selective positive allosteric modulator (PAM) PNU-120596, which were blocked by the α7-selective antagonist methyllycaconitine (MLA). In addition, a small proportion of the neurons express nAChRs with properties typical of heteromeric (non-α7 containing) nAChR subtypes. These cells therefore represent a great tool to advance our understanding of the properties of native human nAChRs, α7 in particular.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Receptors, Nicotinic/genetics , Aconitine/analogs & derivatives , Aconitine/pharmacology , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Isoxazoles/pharmacology , Neurons/metabolism , Phenylurea Compounds/pharmacology , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...