Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 354: 577529, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33676084

ABSTRACT

Peripheral nerve injury frequently evokes chronic neuropathic pain. This is initiated by a transient inflammatory response that leads to persistent excitation of dorsal root ganglion (DRG) neurons by inflammatory cytokines such as interleukin 1ß(IL-1ß). In non-neuronal cells such as lymphocytes, interleukin 1 exerts actions at attomolar (aM; 10-18 M) concentrations. We now report that DRG neurons in defined-medium, neuron-enriched culture display increased excitability following 5-6 d exposure of 1aM IL-1ß. This response is mediated in part by type 1 interleukin receptors and involves decreased function of putative KCa1.1 channels. This finding provides new insights into the neuroimmune interactions responsible for neuropathic pain.


Subject(s)
Ganglia, Spinal/metabolism , Interleukin-1beta/metabolism , Neuralgia/metabolism , Sensory Receptor Cells/metabolism , Animals , Ganglia, Spinal/drug effects , Interleukin-1beta/pharmacology , Mice , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects
2.
Cell Rep ; 33(1): 108233, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33027646

ABSTRACT

The innate immune system responds to infections that give rise to pain. How the innate immune system interacts with the sensory nervous system and contributes to pain is poorly understood. Here we report that hyperactivity of innate immunity primes and initiates pain states via the TLR2-interleukin-33 (IL-33) axis. Toll-like receptors (TLRs) are upregulated in the complete Freund's adjuvant (CFA) pain model, and knockout of TLR2 abolishes CFA-induced pain. Selective activation of TLR2/6 triggers acute pain via upregulation of IL-33 in the hindpaw, dorsal root ganglia (DRG), and spinal cord in an NLRP3-dependent manner. The IL-33 increase further initiates priming of nociceptive neurons and pain states. Finally, blocking IL-33 receptors at the spinal level mediates analgesia during acute and chronic inflammatory pain, underscoring an important function of IL-33 in pain signaling. Collectively, our data reveal a critical role of the TLR2-IL-33 axis in innate immune activation for pain initiation and maintenance.


Subject(s)
Immunity, Innate/genetics , Interleukin-33/metabolism , Toll-Like Receptor 2/metabolism , Animals , Humans , Mice
3.
J Neuroimmunol ; 332: 198-211, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31077855

ABSTRACT

Excitation of dorsal root ganglion (DRG) neurons by interleukin 1ß (IL-1ß) is implicated in the onset of neuropathic pain. To understand its mechanism of action, isolectin B4 positive (IB4+) DRG neurons were exposed to 100pM IL-1ß for 5-6d. A reversible increase in action potential (AP) amplitude reflected increased TTX-sensitive sodium current (TTX-S INa). An irreversible increase in AP duration reflected decreased Ca2+- sensitive K+ conductance (BK(Ca) channels). Different processes thus underlie regulation of the two channel types. Since changes in AP shape facilitated Ca2+ influx, this explains how IL-1ß facilitates synaptic transmission in the dorsal horn; thereby provoking pain.


Subject(s)
Calcium Channels/drug effects , Ganglia, Spinal/cytology , Interleukin-1beta/pharmacology , Ion Channel Gating/drug effects , Neuralgia/etiology , Potassium Channels/drug effects , Sensory Receptor Cells/drug effects , Sodium Channels/drug effects , Action Potentials/drug effects , Animals , Calcium Channels/metabolism , Cell Size , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Male , Nerve Growth Factor/pharmacology , Neuralgia/metabolism , Patch-Clamp Techniques , Peptides/pharmacology , Plant Lectins/analysis , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Sodium Channels/metabolism
4.
Sci Adv ; 4(8): eaas9846, 2018 08.
Article in English | MEDLINE | ID: mdl-30101191

ABSTRACT

Chronic joint pain such as mechanical allodynia is the most debilitating symptom of arthritis, yet effective therapies are lacking. We identify the pannexin-1 (Panx1) channel as a therapeutic target for alleviating mechanical allodynia, a cardinal sign of arthritis. In rats, joint pain caused by intra-articular injection of monosodium iodoacetate (MIA) was associated with spinal adenosine 5'-triphosphate (ATP) release and a microglia-specific up-regulation of P2X7 receptors (P2X7Rs). Blockade of P2X7R or ablation of spinal microglia prevented and reversed mechanical allodynia. P2X7Rs drive Panx1 channel activation, and in rats with mechanical allodynia, Panx1 function was increased in spinal microglia. Specifically, microglial Panx1-mediated release of the proinflammatory cytokine interleukin-1ß (IL-1ß) induced mechanical allodynia in the MIA-injected hindlimb. Intrathecal administration of the Panx1-blocking peptide 10panx suppressed the aberrant discharge of spinal laminae I-II neurons evoked by innocuous mechanical hindpaw stimulation in arthritic rats. Furthermore, mice with a microglia-specific genetic deletion of Panx1 were protected from developing mechanical allodynia. Treatment with probenecid, a clinically used broad-spectrum Panx1 blocker, resulted in a striking attenuation of MIA-induced mechanical allodynia and normalized responses in the dynamic weight-bearing test, without affecting acute nociception. Probenecid reversal of mechanical allodynia was also observed in rats 13 weeks after anterior cruciate ligament transection, a model of posttraumatic osteoarthritis. Thus, Panx1-targeted therapy is a new mechanistic approach for alleviating joint pain.


Subject(s)
Arthralgia/prevention & control , Arthritis, Experimental/prevention & control , Connexins/metabolism , Connexins/physiology , Hyperalgesia/prevention & control , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Spinal Cord Diseases/prevention & control , Animals , Arthralgia/etiology , Arthritis, Experimental/etiology , Connexins/genetics , Hyperalgesia/etiology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley , Spinal Cord Diseases/etiology
5.
Mol Pain ; 13: 1744806917724698, 2017.
Article in English | MEDLINE | ID: mdl-28741432

ABSTRACT

Abstract: We recently reported that nerve injury or peripheral inflammation triggers an upregulation of the deubiquitinase, USP5 in mouse dorsal root ganglion and spinal dorsal horn. This leads to dysregulated ubiquitination of Cav3.2 T-type calcium channels, thus increasing Cav3.2 channel plasma membrane expression and nociceptive signaling in the primary afferent pain pathway. This phenomenon could be recapitulated by noninvasive, optogenetic activation of transient receptor potential vanilloid-1­expressing nociceptors, indicating that neuronal activity is a key player in this process. Given the relevance of the pro-inflammatory cytokine interleukin-1 beta in many forms of pathological pain, we hypothesized that interleukin-1 beta may be a critical cofactor required to drive upregulation of interactions between USP5 and Cav3.2 channels. Here, we report that gene expression, as well as protein levels for interleukin-1 beta and the endogenous interleukin-1 receptor-I antagonist, IL-1Ra are unaltered following conditioning stimulation of optogenetically targeted cutaneous nociceptors, indicating that neuronal activity is not a driver of interleukin-1 beta signaling. In contrast, co-immunoprecipitation experiments revealed that intrathecal administration of interleukin-1 beta in wild-type mice led to an increase in the interaction between USP5 and Cav3.2 in the spinal dorsal horn. Moreover, disruption of the interaction between USP5 and Cav3.2 with TAT peptides suppressed acute nocifensive responses produced by interleukin-1 beta, which was similar to that achieved by elimination of T-type channel activity with the channel blockers, mibefradil, or TTA-A2. Finally, this upregulation could be maintained in dorsal root ganglion neuron cultures exposed overnight to interleukin-1 beta, while the copresence of interleukin-1 receptor antagonist or the dampening of neuronal cell activity with tetrodotoxin attenuated this response. Altogether, our findings identify interleukin-1 beta as an upstream trigger for the upregulation of interactions between USP5 and Cav3.2 channels in the pain pathway, presumably by triggering increased firing activity in afferent fibers.


Subject(s)
Calcium Channels, T-Type/genetics , Interleukin-1beta/metabolism , Pain/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Calcium Channels, T-Type/metabolism , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neuralgia/metabolism , Neurons/metabolism , Nociceptors/metabolism , Up-Regulation
7.
Nat Med ; 23(3): 355-360, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28134928

ABSTRACT

Opiates are essential for treating pain, but termination of opiate therapy can cause a debilitating withdrawal syndrome in chronic users. To alleviate or avoid the aversive symptoms of withdrawal, many of these individuals continue to use opiates. Withdrawal is therefore a key determinant of opiate use in dependent individuals, yet its underlying mechanisms are poorly understood and effective therapies are lacking. Here, we identify the pannexin-1 (Panx1) channel as a therapeutic target in opiate withdrawal. We show that withdrawal from morphine induces long-term synaptic facilitation in lamina I and II neurons within the rodent spinal dorsal horn, a principal site of action for opiate analgesia. Genetic ablation of Panx1 in microglia abolished the spinal synaptic facilitation and ameliorated the sequelae of morphine withdrawal. Panx1 is unique in its permeability to molecules up to 1 kDa in size and its release of ATP. We show that Panx1 activation drives ATP release from microglia during morphine withdrawal and that degrading endogenous spinal ATP by administering apyrase produces a reduction in withdrawal behaviors. Conversely, we found that pharmacological inhibition of ATP breakdown exacerbates withdrawal. Treatment with a Panx1-blocking peptide (10panx) or the clinically used broad-spectrum Panx1 blockers, mefloquine or probenecid, suppressed ATP release and reduced withdrawal severity. Our results demonstrate that Panx1-mediated ATP release from microglia is required for morphine withdrawal in rodents and that blocking Panx1 alleviates the severity of withdrawal without affecting opiate analgesia.


Subject(s)
Behavior, Animal/drug effects , Connexins/genetics , Microglia/drug effects , Morphine/adverse effects , Narcotics/adverse effects , Nerve Tissue Proteins/genetics , Posterior Horn Cells/drug effects , Substance Withdrawal Syndrome/genetics , Adenosine Triphosphate/metabolism , Animals , Apyrase/pharmacology , Blotting, Western , Cell Culture Techniques , Coculture Techniques , Connexins/antagonists & inhibitors , Connexins/metabolism , Mefloquine/pharmacology , Mice , Microglia/metabolism , Naloxone/pharmacology , Narcotic Antagonists/adverse effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Nociception/drug effects , Posterior Horn Cells/metabolism , Probenecid/pharmacology , Rats , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/metabolism
8.
Front Cell Neurosci ; 10: 74, 2016.
Article in English | MEDLINE | ID: mdl-27047338

ABSTRACT

Genetic ablation of cellular prion protein (PrP(C)) has been linked to increased neuronal excitability and synaptic activity in the hippocampus. We have previously shown that synaptic activity in hippocampi of PrP-null mice is increased due to enhanced N-methyl-D-aspartate receptor (NMDAR) function. Here, we focused on the effect of PRNP gene knock-out (KO) on intrinsic neuronal excitability, and in particular, the underlying ionic mechanism in hippocampal neurons cultured from P0 mouse pups. We found that the absence of PrP(C) profoundly affected the firing properties of cultured hippocampal neurons in the presence of synaptic blockers. The membrane impedance was greater in PrP-null neurons, and this difference was abolished by the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker ZD7288 (100 µM). HCN channel activity appeared to be functionally regulated by PrP(C). The amplitude of voltage sag, a characteristic of activating HCN channel current (I h), was decreased in null mice. Moreover, I h peak current was reduced, along with a hyperpolarizing shift in activation gating and slower kinetics. However, neither HCN1 nor HCN2 formed a biochemical complex with PrP(C). These results suggest that the absence of PrP downregulates the activity of HCN channels through activation of a cell signaling pathway rather than through direct interactions. This in turn contributes to an increase in membrane impedance to potentiate neuronal excitability.

9.
Cell Rep ; 12(5): 752-9, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26212331

ABSTRACT

Noxious stimuli are detected by peripheral nociceptors and then transmitted to higher CNS centers, where they are perceived as an unpleasant sensation. The mechanisms that govern the emotional component associated with pain are still incompletely understood. Here, we used optogenetic approaches both in vitro and in vivo to address this issue. We found that peripheral nerve injury inhibits pyramidal cell firing in the prelimbic area of the prefrontal cortex as a result of feed-forward inhibition mediated by parvalbumin-expressing GABAergic interneurons. In addition, activation of inhibitory archaerhodopsin or excitatory channelrhodopsin-2 in these neurons decreased and increased pain responses, respectively, in freely moving mice and accordingly modulated conditioned place preference scores and place escape/avoidance behavior. Our findings thus demonstrate an important role of the prelimbic area in sensory and emotional aspects of pain and identify GABAergic circuits in this region as a potential target for pain therapeutics.


Subject(s)
Avoidance Learning , Behavior, Animal , Emotions , GABAergic Neurons , Neuralgia/physiopathology , Pyramidal Cells , Animals , Male , Mice , Mice, Transgenic , Neuralgia/genetics
10.
J Physiol ; 593(16): 3739-55, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26110238

ABSTRACT

KEY POINTS: Neuropathic pain resulting from peripheral nerve injury is initiated and maintained by persistent ectopic activity in primary afferent neurons. Sciatic nerve injury increases the excitability of medium-sized dorsal root ganglion (DRG) neurons. Levels of the inflammatory cytokine interleukin 1ß (IL-1ß) increase and peak after 7 days. Five to six days of exposure of medium sized DRG neurons to 100 pm IL-1ß promotes persistent increases in excitability which abate within 3-4 days of cytokine removal. This is associated with a profound attenuation of K(+) channel currents but only modest increases in function of cyclic nucleotide-sensitive hyperpolarization-activated channels (HCNs) and of voltage-gated Na(+) and Ca(2+) channel currents. It is unlikely, therefore, that direct interaction of IL-1ß with DRG neurons is capable of initiating an enduring phenotypic shift in their electrophysiological properties that follows sciatic nerve injury. The findings also underline the importance of K(+) channel modulation in the actions of inflammatory mediators on peripheral neurons. ABSTRACT: Chronic constriction injury of rat sciatic nerve promotes signs of neuropathic pain. This is associated with an increase in the level of interleukin 1ß (IL-1ß) in primary afferents that peaks at 7 days. This initial cytokine exposure has been proposed to trigger an enduring alteration in neuronal phenotype that underlies chronic hyper-excitability in sensory nerves, which initiates and maintains chronic neuropathic pain. We have shown previously that 5-6 days of exposure of rat dorsal root ganglia (DRGs) to 100 pm IL-1ß increases the excitability of medium-sized neurons. We have now found using whole-cell recording that this increased excitability reverts to control levels within 3-4 days of cytokine removal. The effects of IL-1ß were dominated by changes in K(+) currents. Thus, the amplitudes of A-current, delayed rectifier and Ca(2+) -sensitive K(+) currents were reduced by ∼68%, ∼64% and ∼36%, respectively. Effects of IL-1ß on other cation currents were modest by comparison. There was thus a slight decrease in availability of high voltage-activated Ca(2+) channel current, a small increase in rates of activation of hyperpolarization-activated cyclic nucleotide-gated channel current (IH ), and a shift in the voltage dependence of activation of tetrodotoxin-sensitive sodium current (TTX-S INa ) to more negative potentials. It is unlikely, therefore, that direct interaction of IL-1ß with DRG neurons initiates an enduring phenotypic shift in their electrophysiological properties following sciatic nerve injury. Persistent increases in primary afferent excitability following nerve injury may instead depend on altered K(+) channel function and on the continued presence of slightly elevated levels IL-1ß and other cytokines.


Subject(s)
Ganglia, Spinal/physiology , Interleukin-1beta/pharmacology , Neurons/drug effects , Potassium Channels/physiology , Animals , Cells, Cultured , Male , Neurons/physiology , Rats, Sprague-Dawley
11.
Neurosci Lett ; 584: 397-402, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25079903

ABSTRACT

The effectiveness of gabapentin (GBP) in the treatment of neuropathic pain depends on access to the α2δ-1 accessory subunit of voltage-gated Ca(2+) channels. Access may be limited by its rate of entry via the neuronal system L-neutral amino acid transporter. The open pore of capsaicin-activated TRPV1 channel admits organic molecules such as local anesthetics and we calculated that GBP entry via this route would be 500× more rapid than via the transporter. Capsaicin should therefore increase GBP effectiveness. We used a quaternary GBP derivative (Q-GBP) as sole charge carrier in whole-cell recording experiments on rat dorsal root ganglion (DRG) neurons. Under these conditions, capsaicin produced a capsazepine-sensitive inward current thereby confirming Q-GBP permeation of TRPV1 channels. We have previously established that 5-6 days exposure to 100 µM GBP decreases excitability of dorsal horn neurons whereas 10 µM is ineffective. Excitability was monitored using confocal Ca(2+) imaging of rat spinal cord slices in organotypic culture. GBP effectiveness was augmented by transient exposures of cultures to capsaicin and robust suppression of excitability was seen with 10 µM GBP. Experiments with an inhibitor of the neutral amino acid transporter, 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH 300 µM), showed the actions of GBP seen in the presence of capsaicin were independent of its entry by this route. Capsaicin potentiation of GBP depression of dorsal horn activity may therefore reflect drug permeation of TRPV1 channels. Agonist activation of TRP channels may provide a means for improving drug access to cytoplasmic targets in selective neuronal populations defined on the basis of type of TRP channel expressed.


Subject(s)
Amines/pharmacology , Analgesics/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Spinal Cord Dorsal Horn/drug effects , TRPV Cation Channels/metabolism , gamma-Aminobutyric Acid/pharmacology , Animals , Calcium/metabolism , Capsaicin/pharmacology , Cells, Cultured , Drug Synergism , Gabapentin , Ganglia, Spinal/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Permeability , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/metabolism
12.
Sci Signal ; 7(346): pe23, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25292211

ABSTRACT

T-type calcium channels are important players in the transmission of pain signals in the primary afferent pathway. Indeed, inhibiting or depleting T-type calcium channels in dorsal root ganglion (DRG) neurons mediates analgesia. Conversely, nerve injury or peripheral inflammation have been shown to induce T-type calcium channel activity in DRG neurons, and this in turn has been linked to the development of chronic pain states. The mechanisms that underlie this enhancement of T-type channels remain incompletely understood and may include changes in channel stability in the plasma membrane or alterations in channel function. In this issue of Science Signaling, Zhang and colleagues identify a cell signaling pathway that potently regulates T-type calcium channel activity in afferent neurons and link this process to pain hypersensitivity. Specifically, they show that insulin-like growth factor-1 receptors in DRG neurons mediate a protein kinase C α (PKCα)-dependent enhancement of T-type calcium currents and that interfering with this pathway reduces both mechanical and thermal pain hypersensitivity in rodents. Targeting this process offers a new avenue for developing pain therapeutics.


Subject(s)
Calcium Channels, T-Type/metabolism , Ganglia, Spinal/metabolism , Insulin-Like Growth Factor I/metabolism , Pain/metabolism , Protein Kinase C-alpha/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Afferent Pathways/metabolism , Animals , Humans , Mice , Models, Neurological
13.
J Neurophysiol ; 112(10): 2398-412, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25122705

ABSTRACT

The α2δ-ligands pregabalin (PGB) and gabapentin (GBP) are used to treat neuropathic pain. We used whole cell recording to study their long-term effects on substantia gelatinosa and dorsal root ganglion (DRG) neurons. Spinal cord slices were prepared from embryonic day 13 rat embryos and maintained in organotypic culture for >5 wk (neuronal age equivalent to young adult rats). Exposure of similarly aged DRG neurons (dissociated and cultured from postnatal day 19 rats) to GBP or PGB for 5-6 days attenuated high-voltage-activated calcium channel currents (HVA ICa). Strong effects were seen in medium-sized and in small isolectin B4-negative (IB4-) DRG neurons, whereas large neurons and small neurons that bound isolectin B4 (IB4+) were hardly affected. GBP (100 µM) or PGB (10 µM) were less effective than 20 µM Mn(2+) in suppression of HVA ICa in small DRG neurons. By contrast, 5-6 days of exposure to these α2δ-ligands was more effective than 20 µM Mn(2+) in reducing spontaneous excitatory postsynaptic currents at synapses in substantia gelatinosa. Spinal actions of gabapentinoids cannot therefore be ascribed to decreased expression of HVA Ca(2+) channels in primary afferent nerve terminals. In substantia gelatinosa, 5-6 days of exposure to PGB was more effective in inhibiting excitatory synaptic drive to putative excitatory neurons than to putative inhibitory neurons. Although spontaneous inhibitory postsynaptic currents were also attenuated, the overall long-term effect of α2δ-ligands was to decrease network excitability as monitored by confocal Ca(2+) imaging. We suggest that selective actions of α2δ-ligands on populations of DRG neurons may predict their selective attenuation of excitatory transmission onto excitatory vs. inhibitory neurons in substantia gelatinosa.


Subject(s)
Amines/pharmacology , Analgesics/pharmacology , Calcium Channel Blockers/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Ganglia, Spinal/drug effects , Substantia Gelatinosa/drug effects , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Excitatory Postsynaptic Potentials/drug effects , Gabapentin , Ganglia, Spinal/physiology , Immunohistochemistry , Inhibitory Postsynaptic Potentials/drug effects , Male , Microscopy, Confocal , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Pregabalin , Rats, Sprague-Dawley , Substantia Gelatinosa/physiology , Tissue Culture Techniques , gamma-Aminobutyric Acid/pharmacology
14.
Can J Neurol Sci ; 39(4): 416-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22728849

ABSTRACT

Neuropathic pain often fails to respond to conventional pain management procedures. here we review the aetiology of neuropathic pain as would result from peripheral neuropathy or injury. We show that inflammatory mediators released from damaged nerves and tissue are responsible for triggering ectopic activity in primary afferents and that this, in turn, provokes increased spinal cord activity and the development of 'central sensitization'. Although evidence is mounting to support the role of interleukin-1ß, prostaglandins and other cytokines in the onset of neuropathic pain, the clinical efficacy of drugs which antagonize or prevent the actions of these mediators is yet to be determined. basic science findings do, however, support the use of pre-emptive analgesia during procedures which involve nerve manipulation and the use of anti-inflammatory steroids as soon as possible following traumatic nerve injury.


Subject(s)
Inflammation/etiology , Ion Channels/physiology , Neuralgia/complications , Neuralgia/pathology , Sensory Receptor Cells/physiology , Animals , Humans
15.
J Neurophysiol ; 107(6): 1586-97, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22170966

ABSTRACT

The effect of interleukin-1ß (IL-1ß) on the electrical properties of sensory neurons was assessed at levels and exposure times comparable to those found in animal models of neuropathic pain. Experiments involved whole cell current-clamp recordings from rat dorsal root ganglion (DRG) neurons in defined-medium, neuron-enriched cultures. Five- to six-day exposure to 100 pM IL-1ß produced subpopulation-dependent effects on DRG neurons. These included an increase in the excitability of medium-diameter and small-diameter isolectin B(4) (IB(4))-positive neurons that was comparable to that found after peripheral nerve injury. By contrast, a reduction in excitability was observed in large-diameter neurons, while no effect was found in small-diameter IB(4)-negative neurons. Further characterization of changes in medium and small IB(4)-positive neurons revealed that some, but not all, effects of IL-1ß were mediated through its receptor, IL-1RI. Although the acute actions of IL-1ß on sensory neurons have been well studied and related to acute and/or inflammatory pain, the present study shows how sensory neurons respond to long-term cytokine exposure. Such effects are relevant to understanding processes that contribute to the onset of neuropathic pain.


Subject(s)
Action Potentials/drug effects , Ganglia, Spinal/drug effects , Interleukin-1beta/pharmacology , Neurons/drug effects , Animals , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley
16.
FASEB J ; 24(11): 4343-53, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20628092

ABSTRACT

Painful peripheral neuropathy has become the principal neurological disorder in HIV/AIDS patients. Herein, we investigated the effects of a cytotoxic HIV-1 accessory protein, viral protein R (Vpr), on the peripheral nervous system (PNS). Host and viral gene expression was investigated in peripheral nerves from HIV-infected individuals and in HIV-infected human dorsal root ganglion (DRG) cultures by RT-PCR and immunocytochemistry. Cytosolic calcium ([Ca(2+)]) fluxes and neuronal membrane responses were analyzed in cultured DRGs. Neurobehavioral responses and cytokine levels were assessed in a transgenic mouse model in which the vpr transgene was expressed in an immunodeficient background (vpr/RAG1(-/-)). Vpr transcripts and proteins were detected in peripheral nerves and DRGs from HIV-infected patients. Exposure of rat or human cultured DRG neurons to Vpr rapidly increased [Ca(2+)] and action potential frequency while increasing input resistance. HIV infection of human DRG cultures caused neurite retraction (P<0.05), accompanied by induction of interferon-α (IFN-α) transcripts (P<0.05). vpr/RAG1(-/-) mice expressed Vpr together with increased IFN-α (P<0.05) in the PNS and also exhibited mechanical allodynia, unlike their vpr/RAG1(-/-) littermates (P<0.05). Herein, Vpr caused DRG neuronal damage, likely through cytosolic calcium activation and cytokine perturbation, highlighting Vpr's contribution to HIV-associated peripheral neuropathy and ensuing neuropathic pain.


Subject(s)
Gene Products, vpr/metabolism , HIV-1 , Neuralgia/complications , Peripheral Nervous System Diseases/complications , Trauma, Nervous System/complications , Animals , Cells, Cultured , Ganglia, Spinal/physiopathology , Ganglia, Spinal/virology , Gene Expression Regulation , Gene Products, vpr/genetics , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Peripheral Nervous System Diseases/physiopathology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
17.
J Pharmacol Exp Ther ; 324(1): 224-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17921188

ABSTRACT

Proteinase-activated receptors (PARs) are a family of G-protein-coupled receptors that are activated by endogenous serine proteinases that cleave the N-terminal domain of the receptor unmasking a "tethered ligand" sequence. Trypsin and other agonists at PAR(2) act on peripheral nerves to augment the transfer of nociceptive information. We tested whether PAR(2) agonists also exert a spinal pronociceptive effect by i.t. administering the selective ligand, Ser-Leu-Ile-Gly-Arg-Leu-NH(2) (SLI-GRL). This produced thermal and mechanical hyperalgesia in rats and mice and augmented mechanical and thermal hyperalgesia seen in the formalin inflammatory pain test. Effects of SLIGRL were abrogated in PAR(2)-deficient mice and were not seen with the inactive control peptide, Leu-Arg-Gly-Ile-Leu-Ser-NH(2). Surprisingly, electrophysiological studies, using whole-cell recording from rat substantia gelatinosa neurons, failed to demonstrate an increase in excitatory transmission or neuronal excitability following treatment with SLIGRL or trypsin. In fact, the actions of trypsin were consistent with a decrease in dorsal horn excitability. SLIGRL and trypsin did, however, depolarize and increase the excitability of large, medium and small primary afferent, dorsal root ganglion neurons. The effects were associated with an increase in conductance at hyperpolarized potentials and a decrease in conductance at depolarized potentials. PAR(2)-like immunoreactivity was found in DRG but not in spinal dorsal horn. These results suggest that activation of DRG neuron cell bodies may account for the pronociceptive actions of i.t. applied PAR(2) agonists. They also imply that pathophysiological release of PAR(2)-activating proteases in the vicinity of DRG neurons may produce profound effects on nociceptive processing in vivo.


Subject(s)
Ganglia, Spinal/cytology , Hyperalgesia/physiopathology , Neurons, Afferent/drug effects , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Animals , Formaldehyde , Ganglia, Spinal/physiology , Hot Temperature , Hyperalgesia/chemically induced , Injections, Spinal , Male , Membrane Potentials/drug effects , Mice , Mice, Knockout , Neurons, Afferent/physiology , Pain/chemically induced , Pain/physiopathology , Rats , Rats, Wistar , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Spinal Cord/drug effects , Spinal Cord/physiopathology , Trypsin/pharmacology
18.
J Neurophysiol ; 96(2): 579-90, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16611846

ABSTRACT

Peripheral nerve injury increases spontaneous action potential discharge in spinal dorsal horn neurons and augments their response to peripheral stimulation. This "central hypersensitivity, " which relates to the onset and persistence of neuropathic pain, reflects spontaneous activity in primary afferent fibers as well as long-term changes in the intrinsic properties of the dorsal horn (centralization). To isolate and investigate cellular mechanisms underlying "centralization," sciatic nerves of 20-day-old rats were subjected to 13-25 days of chronic constriction injury (CCI; Mosconi-Kruger polyethylene cuff model). Spinal cord slices were then acutely prepared from sham-operated or CCI animals, and whole cell recording was used to compare the properties of five types of substantia gelatinosa neuron. These were defined as tonic, irregular, phasic, transient, or delay according to their discharge pattern in response to depolarizing current. CCI did not affect resting membrane potential, rheobase, or input resistance in any neuron type but increased the amplitude and frequency of spontaneous and miniature excitatory postsynaptic currents (EPSCs) in delay, transient, and irregular cells. These changes involved alterations in the action potential-independent neurotransmitter release machinery and possible increases in the postsynaptic effectiveness of glutamate. By contrast, in tonic cells, CCI reduced the amplitude and frequency of spontaneous and miniature EPSCs. Such changes may relate to the putative role of tonic cells as inhibitory GABAergic interneurons, whereas increased synaptic drive to delay cells may relate to their putative role as the excitatory output neurons of the substantia gelatinosa. Complementary changes in synaptic excitation of inhibitory and excitatory neurons may thus contribute to pain centralization.


Subject(s)
Neurons/pathology , Sciatic Nerve/injuries , Substantia Gelatinosa/physiopathology , Animals , Chronic Disease , Constriction, Pathologic/pathology , Constriction, Pathologic/physiopathology , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Membrane Potentials/physiology , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Unmyelinated/physiology , Neurons, Afferent/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sciatic Nerve/pathology , Substantia Gelatinosa/cytology , Substantia Gelatinosa/pathology , Tetrodotoxin/pharmacology
19.
Eur J Neurosci ; 20(11): 2990-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579153

ABSTRACT

Luteinizing hormone releasing hormone (LHRH) is a physiological modulator of neuronal excitability in bullfrog sympathetic ganglia (BFSG). Actions of LHRH involve suppression of the noninactivating, voltage-dependent M-type K+ channel conductance (gM). We found, using whole-cell recordings from these neurons, that LHRH-induced suppression of gM was attenuated by the phospholipase C (PLC) inhibitor U73122 (10 microM) but not by the inactive isomer U73343 (10 microM). Buffering internal Ca2+ to 117 nM with intracellular 20 mM BAPTA + 8 mM Ca2+ or to < 10 nM with intracellular 20 mM BAPTA + 0.4 mM Ca2+ did not attenuate LHRH-induced gM suppression. Suppression of gM by LHRH was not antagonized by the inositol 1,4,5 trisphosphate (InsP3) receptor antagonist heparin (approximately 300 microM). Preventing phosphatidylinositol-4,5-bisphosphate (PIP2) synthesis by blocking phosphatidylinositol-4-kinase with wortmannin (10 microM) or with the nonhydrolysable ATP analogue AMP-PNP (3 mM) prolonged recovery of LHRH-induced gM suppression. This effect was not produced by blocking phosphatidyl inositol-3-kinase with LY294002 (10 microM). Rundown of gM was attenuated when cells were dialysed with 240 microM di-octanoyl PIP2 or 240 microM di-octanoyl phosphatidylinositol-3,4,5-trisphosphate (PIP3) but not with 240 microM di-octanoyl phosphatidylcholine. LHRH-induced gM suppression was competitively antagonized by dialysis with 240 microM di-octanoyl PIP2, but not with di-octanoyl phosphatidylcholine. These results would be expected if LHRH-induced gM suppression reflects a PLC-mediated decrease in plasma membrane PIP2 levels.


Subject(s)
Egtazic Acid/analogs & derivatives , Ganglia, Sympathetic/cytology , Gonadotropin-Releasing Hormone/physiology , Neural Inhibition/physiology , Neurons/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Potassium Channels/metabolism , Animals , Calcium/pharmacology , Cells, Cultured , Chelating Agents/pharmacology , Dialysis/methods , Dose-Response Relationship, Drug , Drug Interactions , Egtazic Acid/pharmacology , Electric Conductivity , Enzyme Inhibitors/pharmacology , Female , Heparin/pharmacology , Intracellular Space/drug effects , Ion Channel Gating/drug effects , Male , Membrane Potentials/drug effects , Neural Inhibition/drug effects , Neural Inhibition/radiation effects , Neurons/drug effects , Neurons/radiation effects , Patch-Clamp Techniques/methods , Potassium Channels/drug effects , Rana catesbeiana/physiology , Time Factors
20.
J Neurosci ; 23(12): 4931-41, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12832515

ABSTRACT

Various neurotransmitters excite neurons by suppressing a ubiquitous, voltage-dependent, noninactivating K+ conductance called the M-conductance (gM). In bullfrog sympathetic ganglion neurons the suppression of gM by the P2Y agonist ATP involves phospholipase C (PLC). The present results are consistent with the involvement of the lipid and inositol phosphate cycles in the effects of both P2Y and muscarinic cholinergic agonists on gM. Impairment of resynthesis of phosphatidylinositol 4,5-bisphosphate (PIP2) with the phosphatidylinositol 4-kinase inhibitor wortmannin (10 microm) slowed or blocked the recovery of agonist-induced gM suppression. This effect could not be attributed to an action of wortmannin on myosin light chain kinase or on phosphatidylinositol 3-kinase. Inhibition of PIP2 synthesis at an earlier point in the lipid cycle by the use of R59022 (40 microm) to inhibit diacylglycerol kinase also slowed the rate of recovery of successive ATP responses. This effect required several applications of agonist to deplete levels of various phospholipid intermediates in the lipid cycle. PIP2 antibodies attenuated the suppression of gM by agonists. Intracellular application of 20 microm PIP2 slowed the rundown of KCNQ2/3 currents expressed in COS-1 or tsA-201 cells, and 100 microm PIP2 produced a small potentiation of native M-current bullfrog sympathetic neurons. These are the results that might be expected if agonist-induced activation of PLC and the concomitant depletion of PIP2 contribute to the excitatory action of neurotransmitters that suppress gM.


Subject(s)
Ganglia, Sympathetic/metabolism , Neurons/metabolism , Neurotransmitter Agents/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels/metabolism , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/metabolism , Adenosine Triphosphate/pharmacology , Aluminum/pharmacology , Androstadienes/pharmacology , Animals , Antibodies/pharmacology , COS Cells , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , KCNQ2 Potassium Channel , KCNQ3 Potassium Channel , Male , Muscarinic Agonists/pharmacology , Neurons/drug effects , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/antagonists & inhibitors , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Potassium Channels/drug effects , Potassium Channels, Voltage-Gated , Rana catesbeiana , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...