Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
FASEB J ; 38(10): e23661, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38733310

ABSTRACT

Itching is an aversive somatosensation that triggers the desire to scratch. Transient receptor potential (TRP) channel proteins are key players in acute and chronic itch. However, whether the modulatory effect of fibroblast growth factor 13 (FGF13) on acute and chronic itch is associated with TRP channel proteins is unclear. Here, we demonstrated that conditional knockout of Fgf13 in dorsal root ganglion neurons induced significant impairment in scratching behaviors in response to acute histamine-dependent and chronic dry skin itch models. Furthermore, FGF13 selectively regulated the function of the TRPV1, but not the TRPA1 channel on Ca2+ imaging and electrophysiological recordings, as demonstrated by a significant reduction in neuronal excitability and current density induced by TRPV1 channel activation, whereas TRPA1 channel activation had no effect. Changes in channel currents were also verified in HEK cell lines. Subsequently, we observed that selective modulation of TRPV1 by FGF13 required its microtubule-stabilizing effect. Furthermore, in FGF13 knockout mice, only the overexpression of FGF13 with a tubulin-binding domain could rescue TRP channel function and the impaired itch behavior. Our findings reveal a novel mechanism by which FGF13 is involved in TRPV1-dependent itch transduction and provide valuable clues for alleviating pathological itch syndrome.


Subject(s)
Fibroblast Growth Factors , Mice, Knockout , Microtubules , Pruritus , TRPV Cation Channels , Animals , Humans , Male , Mice , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Ganglia, Spinal/metabolism , HEK293 Cells , Mice, Inbred C57BL , Microtubules/metabolism , Pruritus/metabolism , Pruritus/genetics , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
2.
Purinergic Signal ; 20(1): 35-45, 2024 Feb.
Article in English | MEDLINE | ID: mdl-36918461

ABSTRACT

Interest has been focused in recent years on the analgesic effects exerted by adenosine and its receptors, A1, A2A, A2B, and A3 adenosine receptor (AR) subtypes, in different in vivo models of chronic pain. In particular, it was demonstrated that selective A3AR agonists reduced pro-nociceptive N-type Ca2+ channels in dorsal root ganglion (DRG) neurons isolated from rats and, by this mechanism, inhibit post inflammatory visceral hypersensitivity. In the present study, we investigate the effect of a previously reported irreversibly binding A3AR agonist, ICBM, on Ca2+ currents (ICa) in rat DRG neurons. Present data demonstrate that ICBM, an isothiocyanate derivative designed for covalent binding to the receptor, concentration-dependently inhibits ICa. This effect is irreversible, since it persists after drug removal, differently from the prototypical A3AR agonist, Cl-IB-MECA. ICBM pre-exposure inhibits the effect of a subsequent Cl-IB-MECA application. Thus, covalent A3AR agonists such as ICBM may represent an innovative, beneficial, and longer-lasting strategy to achieve efficacious chronic pain control versus commonly used, reversible, A3AR agonists. However, the possible limitations of this drug and other covalent drugs may be, for example, a characteristic adverse effect profile, suggesting that more pre-clinical studies are needed.


Subject(s)
Chronic Pain , Ganglia, Spinal , Rats , Animals , Ganglia, Spinal/metabolism , Chronic Pain/metabolism , Neurons/metabolism , Adenosine/metabolism , Receptors, Purinergic P1/metabolism , Receptor, Adenosine A3/metabolism , Adenosine A3 Receptor Agonists/pharmacology
3.
Eur J Neurosci ; 58(6): 3555-3568, 2023 09.
Article in English | MEDLINE | ID: mdl-37608574

ABSTRACT

Limited axon regeneration following peripheral nerve injury may be related to activation of the lysosomal protease, asparaginyl endopeptidase (AEP, δ-secretase) and its degradation of the microtubule associated protein, Tau. Activity of AEP was increased at the site of sciatic nerve transection and repair but blocked in mice treated systemically with a specific AEP inhibitor, compound 11 (CP11). Treatments with CP11 enhanced axon regeneration in vivo. Amplitudes of compound muscle action potentials recorded 4 weeks after nerve transection and repair and 2 weeks after daily treatments with CP11 were double those of vehicle-treated mice. At that time after injury, axons of significantly more motor and sensory neurons had regenerated successfully and reinnervated the tibialis anterior and gastrocnemius muscles in CP11-treated mice than vehicle-treated controls. In cultured adult dorsal root ganglion neurons derived from wild type mice that were treated in vitro for 24 h with CP11, neurites were nearly 50% longer than in vehicle-treated controls and similar to neurite lengths in cultures treated with the TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF). Combined treatment with CP11 and 7,8-DHF did not enhance outgrowth more than treatments with either one alone. Enhanced neurite outgrowth produced by CP11 was found also in the presence of the TrkB inhibitor, ANA-12, indicating that the enhancement was independent of TrkB signalling. Longer neurites were found after CP11 treatment in both TrkB+ and TrkB- neurons. Delta secretase inhibition by CP11 is a treatment for peripheral nerve injury with great potential.


Subject(s)
Axons , Peripheral Nerve Injuries , Animals , Mice , Amyloid Precursor Protein Secretases , Peripheral Nerve Injuries/drug therapy , Nerve Regeneration , Neurites
4.
Brain ; 145(5): 1632-1640, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35661858

ABSTRACT

The axon initial segment is a specialized compartment of the proximal axon of CNS neurons where action potentials are initiated. However, it remains unknown whether this domain is assembled in sensory dorsal root ganglion neurons, in which spikes are initiated in the peripheral terminals. Here we investigate whether sensory neurons have an axon initial segment and if it contributes to spontaneous activity in neuropathic pain. Our results demonstrate that myelinated dorsal root ganglion neurons assemble an axon initial segment in the proximal region of their stem axon, enriched in the voltage-gated sodium channels Nav1.1 and Nav1.7. Using correlative immunofluorescence and calcium imaging, we demonstrate that the Nav1.7 channels at the axon initial segment are associated with spontaneous activity. Computer simulations further indicate that the axon initial segment plays a key role in the initiation of spontaneous discharges by lowering their voltage threshold. Finally, using a Cre-based mouse model for time-controlled axon initial segment disassembly, we demonstrate that this compartment is a major source of spontaneous discharges causing mechanical allodynia in neuropathic pain. Thus, an axon initial segment domain is present in sensory neurons and facilitates their spontaneous activity. This study provides a new insight in the cellular mechanisms that cause pathological pain and identifies a new potential target for chronic pain management.


Subject(s)
Axon Initial Segment , Neuralgia , Animals , Ganglia, Spinal/pathology , Humans , Hyperalgesia/pathology , Mice , Neuralgia/pathology , Sensory Receptor Cells
5.
J Biochem Mol Toxicol ; 37(11): e23459, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37431890

ABSTRACT

Diabetic neuropathy (DN) is the most prevalent complication of diabetes. Pharmacological treatments for DN are often limited in efficacy, so the development of new agents to alleviate DN is essential. The aim of this study was to evaluate the effects of rolipram, a selective phosphodiesterase-4 inhibitor (PDE-4I), and pentoxifylline, a general PDE inhibitor, using a rat model of DN. In this study, a diabetic rat model was established by i.p. injection of STZ (55 mg/kg). Rats were treated with rolipram (1 mg/kg), pentoxifylline (100 mg/kg), and combination of rolipram (0.5 mg/kg) and pentoxifylline (50 mg/kg), orally for 5 weeks. After treatments, sensory function was assessed by hot plate test. Then rats were anesthetized and dorsal root ganglion (DRG) neurons isolated. Cyclic adenosine monophosphate (cAMP), adenosine triphosphate (ATP, adenosine diphosphate and mitochondrial membrane potential (MMP) levels, Cytochrome c release, Bax, Bcl-2, caspase-3 proteins expression in DRG neurons were assessed by biochemical and ELISA methods, and western blot analysis. DRG neurons were histologically examined using hematoxylin and eosin (H&E) staining method. Rolipram and/or pentoxifylline significantly attenuated sensory dysfunction by modulating nociceptive threshold. Rolipram and/or pentoxifylline treatment dramatically increased the cAMP level, prevented mitochondrial dysfunction, apoptosis and degeneration of DRG neurons, which appears to be mediated by inducing ATP and MMP, improving cytochrome c release, as well as regulating the expression of Bax, Bcl-2, and caspase-3 proteins, and improving morphological abnormalities of DRG neurons. We found maximum effectiveness with rolipram and pentoxifylline combination on mentioned factors. These findings encourage the use of rolipram and pentoxifylline combination as a novel experimental evidence for further clinical investigations in the treatment of DN.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Pentoxifylline , Rats , Animals , Pentoxifylline/pharmacology , Pentoxifylline/therapeutic use , Rolipram/pharmacology , Rolipram/metabolism , Rolipram/therapeutic use , Diabetic Neuropathies/metabolism , Caspase 3/metabolism , Cytochromes c/metabolism , Ganglia, Spinal/metabolism , bcl-2-Associated X Protein/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Apoptosis , Neurons/metabolism , Adenosine Triphosphate/metabolism , Mitochondria , Diabetes Mellitus/metabolism
6.
Acta Pharmacol Sin ; 44(9): 1768-1776, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37142682

ABSTRACT

Voltage-gated sodium channel 1.7 (Nav1.7) remains one of the most promising drug targets for pain relief. In the current study, we conducted a high-throughput screening of natural products in our in-house compound library to discover novel Nav1.7 inhibitors, then characterized their pharmacological properties. We identified 25 naphthylisoquinoline alkaloids (NIQs) from Ancistrocladus tectorius to be a novel type of Nav1.7 channel inhibitors. Their stereostructures including the linkage modes of the naphthalene group at the isoquinoline core were revealed by a comprehensive analysis of HRESIMS, 1D, and 2D NMR spectra as well as ECD spectra and single-crystal X-ray diffraction analysis with Cu Kα radiation. All the NIQs showed inhibitory activities against the Nav1.7 channel stably expressed in HEK293 cells, and the naphthalene ring in the C-7 position displayed a more important role in the inhibitory activity than that in the C-5 site. Among the NIQs tested, compound 2 was the most potent with an IC50 of 0.73 ± 0.03 µM. We demonstrated that compound 2 (3 µM) caused dramatical shift of steady-state slow inactivation toward the hyperpolarizing direction (V1/2 values were changed from -39.54 ± 2.77 mV to -65.53 ± 4.39 mV, which might contribute to the inhibition of compound 2 against the Nav1.7 channel. In acutely isolated dorsal root ganglion (DRG) neurons, compound 2 (10 µM) dramatically suppressed native sodium currents and action potential firing. In the formalin-induced mouse inflammatory pain model, local intraplantar administration of compound 2 (2, 20, 200 nmol) dose-dependently attenuated the nociceptive behaviors. In summary, NIQs represent a new type of Nav1.7 channel inhibitors and may act as structural templates for the following analgesic drug development.


Subject(s)
Alkaloids , NAV1.7 Voltage-Gated Sodium Channel , Mice , Animals , Humans , HEK293 Cells , Pain/drug therapy , Neurons , Alkaloids/pharmacology , Alkaloids/therapeutic use , Ganglia, Spinal , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use
7.
Int J Toxicol ; 42(1): 4-18, 2023.
Article in English | MEDLINE | ID: mdl-36308016

ABSTRACT

Previous studies using monotypic nerve cell cultures have shown that nanoparticles induced neurotoxic effects on nerve cells. Interactions between neurons and Schwann cells may protect against the neurotoxicity of nanoparticles. In this study, we developed a co-culture model consisting of immortalized rat dorsal root ganglion (DRG) neurons and rat Schwann cells and employed it to investigate our hypothesis that co-culturing DRG neurons with Schwann cells imparts protection on them against neurotoxicity induced by silver or gold nanoparticles. Our results indicated that neurons survived better in co-cultures when they were exposed to these nanoparticles at the higher concentrations compared to when they were exposed to these nanoparticles at the same concentrations in monotypic cultures. Synapsin I expression was increased in DRG neurons when they were co-cultured with Schwann cells and treated with or without nanoparticles. Glial fibrillary acidic protein (GFAP) expression was increased in Schwann cells when they were co-cultured with DRG neurons and treated with nanoparticles. Furthermore, we found co-culturing with Schwann cells stimulated neurofilament polymerization in DRG neurons and produced the morphological differentiation. Silver nanoparticles induced morphological disorganization in monotypic cultures. However, there were more cells displaying normal morphology in co-cultures than in monotypic cultures. All of these results suggested that co-culturing DRG neurons with Schwann cells imparted some protection on them against neurotoxicity induced by silver or gold nanoparticles, and altering the expression of neurofilament-L, synapsin I, and GFAP could account for the phenomenon of protection in co-cultures.


Subject(s)
Coculture Techniques , Metal Nanoparticles , Neurons , Animals , Rats , Cells, Cultured , Coculture Techniques/methods , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gold/toxicity , Metal Nanoparticles/toxicity , Neurons/metabolism , Schwann Cells/metabolism , Silver/toxicity , Synapsins/pharmacology
8.
Inflamm Res ; 71(4): 485-495, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35298670

ABSTRACT

OBJECTIVE: The transient receptor potential vanilloid subtype 1 (TRPV1) channel is considered to play an important regulatory role in the process of pain. The purpose of this study is to observe the change characteristics of TRPV1 channel in MSU-induced gouty arthritis and to find a new target for clinical treatment of gout pain. METHODS: Acute gouty arthritis was induced by injection of monosodium urate (MSU) crystals into the ankle joint of mice. The swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical hyperalgesia was conducted using the electronic von Frey. Calcium fluorescence and TRPV1 current were recorded by applying laser scanning confocal microscope and patch clamp in dorsal root ganglion (DRG) neurons, respectively. RESULTS: MSU treatment resulted in significant inflammatory response and mechanical hyperalgesia. The peak swelling degree appeared at 12 h, and the minimum pain threshold appeared at 8 h after MSU treatment. The fluorescence intensity of capsaicin-induced calcium response and TRPV1 current were increased in DRG cells from MSU-treated mice. The number of cells that increased calcium response after MSU treatment was mainly distributed in small-diameter DRG cells. However, the action potential was not significantly changed in small-diameter DRG cells after MSU treatment. CONCLUSIONS: These findings identified an important role of TRPV1 in mediating mechanical hyperalgesia in MSU-induced gouty arthritis and further suggest that TRPV1 can be regarded as a potential new target for the clinical treatment of gouty arthritis.


Subject(s)
Arthritis, Gouty , Transient Receptor Potential Channels , Animals , Arthritis, Gouty/chemically induced , Arthritis, Gouty/drug therapy , Calcium , Edema , Hyperalgesia/chemically induced , Mice , Pain , TRPV Cation Channels , Transient Receptor Potential Channels/therapeutic use , Uric Acid
9.
Metab Brain Dis ; 37(7): 2615-2627, 2022 10.
Article in English | MEDLINE | ID: mdl-35922732

ABSTRACT

Diabetic neuropathy (DN) is the most challenging microvascular complication of diabetes and there is no suitable treatment for it, so the development of new agents to relieve DN is urgently needed. Since oxidative stress and inflammation play an essential role in the development of DN, clearance of these factors are good strategies for the treatment of this disease. According to key role of cyclic adenosine monophosphate (cAMP) in the regulation of oxidative stress and inflammatory pathways, it seems that phosphodiesterase inhibitors (PDEIs) can be as novel drug targets for improving DN through enhancement of cAMP level. The aim of this study was to evaluate the effects of rolipram, a selective PDE4 inhibitor, and pentoxifylline, a general PDE inhibitor on experimental model of DN and also to determine the possible mechanisms involved in the effectiveness of these agents. We investigated the effects of rolipram (1 mg/kg) and pentoxifylline (100 mg/kg) and also combination of rolipram (0.5 mg/kg) and pentoxifylline (50 mg/kg), orally for five weeks in rats that became diabetic by STZ (55 mg/kg, i.p.). After treatments, motor function was evaluated by open-field test, then rats were anesthetized and dorsal root ganglion (DRG) neurons isolated. Next, oxidative stress biomarkers and inflammatory factors were assessed by biochemical and ELISA methods, and RT-PCR analysis in DRG neurons. Rolipram and/or pentoxifylline treatment significantly attenuated DN - induced motor function deficiency by modulating distance moved and velocity. Rolipram and/or pentoxifylline treatment dramatically increased the cAMP level, as well as suppressed DN - induced oxidative stress which was associated with decrease in LPO and ROS and increase in TAC, total thiol, CAT and SOD in DRG neurons. On the other hand, the level of inflammatory factors (TNF-α, NF-kB and COX2) significantly decreased following rolipram and/or pentoxifylline administration. The maximum effectiveness was with rolipram and/or pentoxifylline combination on mentioned factors. These findings provide novel experimental evidence for further clinical investigations on rolipram and pentoxifylline combination for the treatment of DN.


Subject(s)
Diabetic Neuropathies , Pentoxifylline , Phosphodiesterase 4 Inhibitors , Animals , Rats , Rolipram/pharmacology , Rolipram/therapeutic use , Pentoxifylline/pharmacology , Pentoxifylline/therapeutic use , Diabetic Neuropathies/drug therapy , Ganglia, Spinal/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species , NF-kappa B/metabolism , Cyclooxygenase 2/metabolism , Oxidative Stress , Neurons/metabolism , Biomarkers/metabolism , Sulfhydryl Compounds , Adenosine Monophosphate/metabolism , Superoxide Dismutase/metabolism
10.
Int J Mol Sci ; 23(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36077386

ABSTRACT

Oxaliplatin (OHP) is a platinum-based agent that can cause peripheral neuropathy, an adverse effect in which the dorsal root ganglion (DRG) neurons are targeted. Zonisamide has exhibited neuroprotective activities toward adult rat DRG neurons in vitro and therefore, we aimed to assess its potential efficacy against OHP-induced neurotoxicity. Pretreatment with zonisamide (100 µM) alleviated the DRG neuronal death caused by OHP (75 µM) and the protective effects were attenuated by a co-incubation with 25 µM of the mitogen-activated protein kinase (MAPK; MEK/ERK) inhibitor, U0126, or the phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor, LY294002. Pretreatment with zonisamide also suppressed the OHP-induced p38 MAPK phosphorylation in lined DRG neurons, ND7/23, while the OHP-induced DRG neuronal death was alleviated by pretreatment with the p38 MAPK inhibitor, SB239063 (25 µM). Although zonisamide failed to protect the immortalized rat Schwann cells IFRS1 from OHP-induced cell death, it prevented neurite degeneration and demyelination-like changes, as well as the reduction of the serine/threonine-specific protein kinase (AKT) phosphorylation in DRG neuron-IFRS1 co-cultures exposed to OHP. Zonisamide's neuroprotection against the OHP-induced peripheral sensory neuropathy is possibly mediated by a stimulation of the MEK/ERK and PI3K/AKT signaling pathways and suppression of the p38 MAPK pathway in DRG neurons. Future studies will allow us to solidify zonisamide as a promising remedy against the neurotoxic adverse effects of OHP.


Subject(s)
Ganglia, Spinal , Peripheral Nervous System Diseases , Animals , Cells, Cultured , Coculture Techniques , Ganglia, Spinal/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/metabolism , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/chemically induced , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Schwann Cells/metabolism , Zonisamide/adverse effects , Zonisamide/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Molecules ; 27(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35335254

ABSTRACT

Ligands of the Gi protein-coupled adenosine A3 receptor (A3R) are receiving increasing interest as attractive therapeutic tools for the treatment of a number of pathological conditions of the central and peripheral nervous systems (CNS and PNS, respectively). Their safe pharmacological profiles emerging from clinical trials on different pathologies (e.g., rheumatoid arthritis, psoriasis and fatty liver diseases) confer a realistic translational potential to these compounds, thus encouraging the investigation of highly selective agonists and antagonists of A3R. The present review summarizes information on the effect of latest-generation A3R ligands, not yet available in commerce, obtained by using different in vitro and in vivo models of various PNS- or CNS-related disorders. This review places particular focus on brain ischemia insults and colitis, where the prototypical A3R agonist, Cl-IB-MECA, and antagonist, MRS1523, have been used in research studies as reference compounds to explore the effects of latest-generation ligands on this receptor. The advantages and weaknesses of these compounds in terms of therapeutic potential are discussed.


Subject(s)
Adenosine A3 Receptor Agonists , Arthritis, Rheumatoid , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/therapeutic use , Arthritis, Rheumatoid/drug therapy , Humans , Ligands , Peripheral Nervous System , Receptors, Purinergic P1
12.
Clin Exp Pharmacol Physiol ; 48(3): 347-354, 2021 03.
Article in English | MEDLINE | ID: mdl-33064853

ABSTRACT

The voltage-gated sodium channel (VGSC) currents in dorsal root ganglion (DRG) neurons contain mainly TTX-sensitive (TTX-S) and TTX-resistant (TTX-R) Na+ currents. Magnolol (Mag), a hydroxylated biphenyl compound isolated from the bark of Magnolia officinalis, has been well documented to exhibit analgesic effects, but its mechanism is not yet fully understood. The aim of the present study was to investigate whether the antinociceptive effects of Mag is through inhibition of Na+ currents. Na+ currents in freshly isolated mouse DRG neurons were recorded with the whole cell patch clamp technique. Results showed that Mag inhibited TTX-S and TTX-R Na+ currents in a concentration-dependent manner. The IC50 values for block of TTX-S and TTX-R Na+ currents were 9.4 and 7.0 µmol/L, respectively. Therefore, TTX-R Na+ current was more susceptible to Mag than TTX-S Na+ current. For TTX-S Na+ channel, 10 µmol/L Mag shifted the steady state inactivation curve toward more negative by 9.8 mV, without affecting the activation curve. For TTX-R Na+ channel, 7 µmol/L Mag shifted the steady state activation and inactivation curves toward more positive and negative potentials by 6.5 and 11.7 mV, respectively. In addition, Mag significantly postponed recovery of TTX-S and TTX-R Na+ currents from inactivation, and produced frequency dependent blocks of both subtypes of Na+ currents. These results suggest that the inhibitory effects of Mag on Na+ channels may contribute to its analgesic effect.


Subject(s)
Biphenyl Compounds , Ganglia, Spinal , Lignans , Sodium , Patch-Clamp Techniques , Tetrodotoxin
13.
Int J Mol Sci ; 22(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34360719

ABSTRACT

Agonists of the Gi protein-coupled A3 adenosine receptor (A3AR) have shown important pain-relieving properties in preclinical settings of several pain models. Active as a monotherapy against chronic pain, A3AR agonists can also be used in combination with classic opioid analgesics. Their safe pharmacological profile, as shown by clinical trials for other pathologies, i.e., rheumatoid arthritis, psoriasis and fatty liver diseases, confers a realistic translational potential, thus encouraging research studies on the molecular mechanisms underpinning their antinociceptive actions. A number of pathways, involving central and peripheral mechanisms, have been proposed. Recent evidence showed that the prototypical A3AR agonist Cl-IB-MECA and the new, highly selective, A3AR agonist MRS5980 inhibit neuronal (N-type) voltage-dependent Ca2+ currents in dorsal root ganglia, a known pain-related mechanism. Other proposed pathways involve reduced cytokine production, immune cell-mediated responses, as well as reduced microglia and astrocyte activation in the spinal cord. The aim of this review is to summarize up-to-date information on A3AR in the context of pain, including cellular and molecular mechanisms underlying this effect. Based on their safety profile shown in clinical trials for other pathologies, A3AR agonists are proposed as novel, promising non-narcotic agents for pain control.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Calcium Signaling/drug effects , Ganglia, Spinal , Pain , Receptor, Adenosine A3/metabolism , Animals , Astrocytes/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Humans , Microglia/metabolism , Pain/drug therapy , Pain/metabolism , Pain/physiopathology
14.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804063

ABSTRACT

Besides its insulinotropic actions on pancreatic ß cells, neuroprotective activities of glucagon-like peptide-1 (GLP-1) have attracted attention. The efficacy of a GLP-1 receptor (GLP-1R) agonist exendin-4 (Ex-4) for functional repair after sciatic nerve injury and amelioration of diabetic peripheral neuropathy (DPN) has been reported; however, the underlying mechanisms remain unclear. In this study, the bioactivities of Ex-4 on immortalized adult rat Schwann cells IFRS1 and adult rat dorsal root ganglion (DRG) neuron-IFRS1 co-culture system were investigated. Localization of GLP-1R in both DRG neurons and IFRS1 cells were confirmed using knockout-validated monoclonal Mab7F38 antibody. Treatment with 100 nM Ex-4 significantly enhanced survival/proliferation and migration of IFRS1 cells, as well as stimulated the movement of IFRS1 cells toward neurites emerging from DRG neuron cell bodies in the co-culture with the upregulation of myelin protein 22 and myelin protein zero. Because Ex-4 induced phosphorylation of serine/threonine-specific protein kinase AKT in these cells and its effects on DRG neurons and IFRS1 cells were attenuated by phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor LY294002, Ex-4 might act on both cells to activate PI3K/AKT signaling pathway, thereby promoting myelination in the co-culture. These findings imply the potential efficacy of Ex-4 toward DPN and other peripheral nerve lesions.


Subject(s)
Diabetic Neuropathies/drug therapy , Exenatide/pharmacology , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Animals , Cell Movement/genetics , Cell Survival/genetics , Chromones/pharmacology , Coculture Techniques , Diabetic Neuropathies/genetics , Diabetic Neuropathies/pathology , Exenatide/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Morpholines/pharmacology , Myelin Sheath/genetics , Myelin Sheath/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Schwann Cells/cytology , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/injuries
15.
Inflammopharmacology ; 29(3): 869-877, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34021831

ABSTRACT

Voltage-gated sodium channels are currently recognized as one of the targets of analgesics. Magnolol (Mag), an active component isolated from Magnolia officinalis, has been reported to exhibit analgesic effects. The objective of this study was to investigate whether the analgesic effect of Mag was associated with blocking Na+ channels. Inflammatory pain was induced by the injection of carrageenan into the hind paw of mice. Mag was administered orally. Mechanical hyperanalgesia was evaluated by using von Frey filaments. Na+ currents and neuronal excitability in acutely isolated mouse dorsal root ganglion (DRG) neurons were recorded with the whole-cell patch clamp technique. Results showed that Mag (10 ~ 40 mg/kg) dose-dependently inhibited the paw edema and reduced mechanical pain in the inflammatory animal model. Injection of carrageenan significantly increased the amplitudes of TTX-sensitive and TTX-resistant Na+ currents. Compared with the carrageenan group, Mag inhibited the upregulation of two types of Na+ currents induced by carrageenan in a dose-dependent manner. Mag 40 mg/kg shifted the inactivation curves of two types of Na+ currents to hyperpolarization and returned to normal animal level without changing their activation curves. Mag 40 mg/kg significantly reduced the percentage of cells firing multiple spikes and inhibited the neuronal hyperexcitability induced by carrageenan. Our data suggest that the analgesic effect of Mag may be associated with a decreased neuronal excitability by blocking Na+ current.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biphenyl Compounds/therapeutic use , Ganglia, Spinal/drug effects , Lignans/therapeutic use , Neurons/drug effects , Pain/drug therapy , Sodium Channel Blockers/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biphenyl Compounds/pharmacology , Carrageenan/toxicity , Cells, Cultured , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Edema/metabolism , Ganglia, Spinal/physiology , Lignans/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Neurons/physiology , Pain/physiopathology , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology
16.
J Cell Physiol ; 235(6): 5192-5203, 2020 06.
Article in English | MEDLINE | ID: mdl-31729029

ABSTRACT

The transient receptor potential melastatin type 8 (TRPM8) receptor channel is expressed in primary afferent neurons where it is the main transducer of innocuous cold temperatures and also in a variety of tumors, where it is involved in progression and metastasis. Modulation of this channel by intracellular signaling pathways has therefore important clinical implications. We investigated the modulation of recombinant and natively expressed TRPM8 by the Src kinase, which is known to be involved in cancer pathophysiology and inflammation. Human TRPM8 expressed in HEK293T cells is constitutively tyrosine phosphorylated by Src which is expressed either heterologously or endogenously. Src action on TRPM8 potentiates its activity, as treatment with PP2, a selective Src kinase inhibitor, reduces both TRPM8 tyrosine phosphorylation and cold-induced channel activation. RNA interference directed against the Src kinase diminished the extent of PP2-induced functional downregulation of TRPM8, confirming that PP2 acts mainly through Src inhibition. Finally, the effect of PP2 on TRPM8 cold activation was reproduced in cultured rat dorsal root ganglion neurons, and this action was antagonized by the protein tyrosine phosphatase inhibitor pervanadate, confirming that TRPM8 activity is sensitive to the cellular balance between tyrosine kinases and phosphatases. This positive modulation of TRPM8 by Src kinase may be relevant for inflammatory pain and cancer signaling.


Subject(s)
Inflammation/genetics , Neurons, Afferent/metabolism , TRPM Cation Channels/genetics , src-Family Kinases/genetics , Animals , Biological Transport/genetics , Cold Temperature , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/pathology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Neurons, Afferent/pathology , Pain/drug therapy , Pain/genetics , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Rats , Tyrosine/metabolism , src-Family Kinases/antagonists & inhibitors
17.
J Cell Biochem ; 121(2): 1790-1800, 2020 02.
Article in English | MEDLINE | ID: mdl-31642099

ABSTRACT

To search the modulation mechanism of loureirin B, a flavonoid is extracted from Dracaena cochinchinensis, on tetrodotoxin-resistant (TTX-R) sodium channel in dorsal root ganglion (DRG) neurons of rats. Experiments were carried out based on patch-clamp technique and molecular biological methods. We observed the time-dependent inhibition of loureirin B on TTX-R sodium currents in DRG neurons and found that neither occupancy theory nor rate theory could well explain the time-dependent inhibitory effect of loureirin B on TTX-R sodium currents. It suggested that a second messenger-mediated signaling pathway may be involved in the modulation mechanism. So the cyclin AMP (cAMP) level of the DRG neurons before and after incubation with loureirin B was tested by ELISA Kit. Results showed that loureirin B could increase the cAMP level and the increased cAMP was caused by the enhancement of adenylate cyclase (AC) induced by loureirin B. Immunolabelling experiments further confirmed that loureirin B can promote the production of PKA in DRG neurons. In the presence of the PKA inhibitor H-89, the inhibitory effect of loureirin B on TTX-R sodium currents was reversed. Forskolin, a tool in biochemistry to raise the levels of cAMP, also could reduce TTX-R sodium currents similar to that of loureirin B. These studies demonstrated that loureirin B can modulate the TTX-R sodium channel in DRG neurons via an AC/cAMP/PKA pathway involving the activation of AC and PKA, which also can be used to explain the other pharmacological effects of loureirin B.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Resistance , Ganglia, Spinal/physiology , Neurons/physiology , Resins, Plant/pharmacology , Sodium Channels/chemistry , Tetrodotoxin/pharmacology , Action Potentials , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Membrane Potentials , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology
18.
Cell Physiol Biochem ; 54(4): 577-590, 2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32506846

ABSTRACT

BACKGROUND/AIMS: Loss-of-Function (LOF) of the potassium chloride cotransporter 3 (KCC3) results in hereditary sensorimotor neuropathy with Agenesis of the Corpus Callosum (HSMN/ACC). Our KCC3 knockout mouse recapitulated axonal swelling and tissue vacuolization observed in autopsies of individuals with HSMN/ACC. We previously documented the first human case of a KCC3 gain-of-function (GOF) in which the patient also exhibited severe peripheral neuropathy. Furthermore, the GOF mouse model exhibited shrunken axons implicating the cotransporter in cell volume homeostasis. It is unclear how both KCC3 LOF and GOF lead to peripheral neuropathy. Thus, we sought to study differences in cell volume regulation of dorsal root ganglion neurons isolated from different mouse lines. METHODS: Using wide-field microscopy, we measured calcein fluorescence intensity through pinhole measurements at the center of cells and compared cell swelling and cell volume regulation/recovery of wild-type, LOF, and GOF dorsal root ganglia neurons, as well as wild-type neurons treated with a KCC-specific inhibitor. RESULTS: In contrast to control neurons that swell and volume regulate under a hypotonic challenge, neurons lacking KCC3 swell but fail to volume regulate. Similar data were observed in wild-type neurons treated with the KCC inhibitor. We also show that sensory neurons expressing a constitutively active KCC3 exhibited a blunted swelling phase compared to wild-type neurons, questioning the purely osmotic nature of the swelling phase. CONCLUSION: These findings demonstrate the integral role of KCC3 in cell volume homeostasis and support the idea that cell volume homeostasis is critical to the health of peripheral nerves.


Subject(s)
Corpus Callosum/metabolism , Ganglia, Spinal/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Membrane Transport Proteins/metabolism , Neurons/metabolism , Symporters/metabolism , Animals , Axons/metabolism , Cell Size/drug effects , Corpus Callosum/pathology , Disease Models, Animal , Gain of Function Mutation , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hereditary Sensory and Autonomic Neuropathies/genetics , Homeostasis , Humans , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Osmotic Pressure/physiology , Symporters/antagonists & inhibitors , Symporters/genetics
19.
Histochem Cell Biol ; 153(3): 177-184, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31879799

ABSTRACT

Zonisamide, an anti-epileptic and anti-Parkinson's disease drug, displays neurotrophic activity on cultured motor neurons and facilitates axonal regeneration after peripheral nerve injury in mice, but its underlying mechanisms remain unclear. In this study, zonisamide enhanced neurite outgrowth from cultured adult rat dorsal root ganglion (DRG) neurons in a concentration-dependent manner (1 µM < 10 µM < 100 µM), and its activity was significantly attenuated by co-treatment with a phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor LY294002 or a mitogen-activated protein kinase (MAPK) inhibitor U0126. In agreement with these findings, 100 µM zonisamide for 1 h induced phosphorylation of AKT and ERK1/2, key molecules of PI3K and MAPK signaling pathways, respectively in mouse neuroblastoma × rat DRG neuron hybrid cells ND7/23. In contrast, zonisamide failed to promote proliferation or migration of immortalized Fischer rat Schwann cells 1 (IFRS1). These findings suggest that the beneficial effects of zonisamide on peripheral nerve regeneration may be attributable to its direct actions on neurons through PI3K and MAPK pathways, rather than the stimulation of Schwann cells.


Subject(s)
Anticonvulsants/pharmacology , Ganglia, Spinal/drug effects , Neurites/drug effects , Neuronal Outgrowth/drug effects , Neurons/drug effects , Zonisamide/pharmacology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/metabolism , Neurites/metabolism , Neurons/metabolism , Rats , Rats, Wistar , Schwann Cells/cytology , Schwann Cells/metabolism , Structure-Activity Relationship
20.
Inflamm Res ; 69(6): 589-598, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32306120

ABSTRACT

OBJECTIVE: To evaluate the role of K+ channels in pain following gouty arthritis. METHODS: The model of acute gouty arthritis was induced by monosodium urate (MSU) in mice. The swelling degree was determined by measuring the circumference of the ankle joint. Mechanical hyperalgesia was detected by von Frey filaments. Two types of K+ currents, A-type currents (IA) and delayed rectifier currents (IK), were recorded in dorsal root ganglion (DRG) neurons using patch-clamp techniques. RESULTS: The swelling degree reached its maximum at 10 h and the minimum pain threshold was maintained between 8 and 48 h after MSU treatment in mice. The amplitudes of IA and IK in DRG neurons were moderately increased on day 1 after MSU treatment, and then, they were gradually decreased with times and reached their minimums on day 4 (for IA) or 5 (for IK). Compared with control group, the activation curve of IA was significantly shifted to more positive potential and the recovery time of IA from inactivation was markedly prolonged, but inactivation and frequency dependence of IA appeared unaffected in MSU-treated group. Additionally, no change was observed in the activation curve of IK after MSU treatment. The excitability was significantly higher in the MSU group than in the control group. CONCLUSIONS: MSU-induced gout pain may be related to the hyperexcitability of DRG neurons elicited by decreasing K+ currents.


Subject(s)
Arthritis, Gouty/physiopathology , Pain/physiopathology , Potassium Channels, Voltage-Gated/physiology , Animals , Arthritis, Gouty/chemically induced , Ganglia, Spinal/physiology , Male , Mice, Inbred ICR , Neurons/physiology , Pain/chemically induced , Uric Acid
SELECTION OF CITATIONS
SEARCH DETAIL