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










Publication year range
1.
Mol Pain ; 15: 1744806919847810, 2019.
Article in English | MEDLINE | ID: mdl-30983496

ABSTRACT

Electroacupuncture has been shown to effectively reduce chronic pain in patients with nerve injury. The underlying mechanisms are not well understood. Accumulated evidence suggests that purinergic P2X3 receptors (P2X3Rs) in dorsal root ganglion neurons play a major role in mediating chronic pain associated with nerve injury. The aim of this study is to determine if electroacupuncture stimulation alters P2X3R activity in dorsal root ganglia to produce analgesia under neuropathic pain condition. Peripheral neuropathy was produced by ligation of the left lumbar 5 (L5) spinal nerve in rats. Low-frequency (2 Hz) electrical stimulation was applied to ipsilateral ST36 and BL60 acupoints in rats. The P2X3R agonist (α,ß-meATP)-induced flinch responses were reduced after electroacupuncture treatment. Western analyses showed that P2X3R expression was upregulated in nerve-uninjured lumbar 4 (L4) dorsal root ganglion neurons ipsilateral to the spinal nerve ligation. Electroacupuncture-stimulation reversed the upregulation. In nerve-injured L5 dorsal root ganglia, P2X3R expression was substantially reduced. Electroacupuncture had no effect on the reduction. We also determined the injury state of P2X3R expressing dorsal root ganglion neurons using the neuronal injury marker, activating transcription factor 3 (ATF3). Immunohistochemical assay showed that in L4 dorsal root ganglia, almost all P2X3Rs were expressed in uninjured (ATF3-) neurons. Spinal nerve ligation increased the expression of P2X3Rs. Electroacupuncture reduced the increase in P2X3R expression without affecting the percentage of ATF + neurons. In ipsilateral L5 dorsal root ganglion neurons, spinal nerve ligation reduced the percentage of P2X3R + neurons and markedly increased the percentage of ATF3 + cells. Almost all of P2X3Rs were expressed in damaged (ATF3+) neurons. Electroacupuncture had no effect on spinal nerve ligation-induced changes in the percentage of P2X3R or percentage of ATF3 + cells in L5 dorsal root ganglia. These observations led us to conclude that electroacupuncture effectively reduces injury-induced chronic pain by selectively reducing the expression of P2X3Rs in nerve-uninjured L4 dorsal root ganglion neurons.


Subject(s)
Down-Regulation , Electroacupuncture , Ganglia, Spinal/metabolism , Receptors, Purinergic P2X3/metabolism , Spinal Nerves/metabolism , Activating Transcription Factor 3/metabolism , Adenosine Triphosphate/analogs & derivatives , Animals , Ganglia, Spinal/pathology , Hyperalgesia/pathology , Ligation , Lumbar Vertebrae/pathology , Male , Neurons/pathology , Rats, Sprague-Dawley
2.
Pain ; 159(11): 2383-2393, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30015706

ABSTRACT

The exchange proteins activated by cAMP (Epacs) have been shown to play important roles in producing inflammation-induced nociception. Transient receptor potential vanilloid type 1 (TRPV1) is a major receptor processing thermal and chemosensitive nociceptive information. The role of Epacs in modulating the activity of TRPV1 has yet to be determined. Studying the effect of complete Freund adjuvant (CFA)-induced inflammation on capsaicin-activated TRPV1 nociceptive responses in dorsal root ganglia (DRG), we found that CFA produced a large increase in capsaicin-induced responses. The increase was inhibited by Epac1 and Epac2 antagonists. Thus, activation of Epacs is critical in producing enhancement in TRPV1-mediated responses under inflammatory conditions. In addition, the inflammation-induced enhancement of TRPV1 responses was blocked by PKCα and PKCε inhibitors, suggesting the essential roles of these PKCs in enhancing TRPV1 responses. To determine the mechanism underlying the Epac actions on TRPV1, we studied the effects of the Epac activator, 8-(4-chlorophenylthio)-2-O-methyl-cAMP (CPT), on capsaicin-induced nociceptive behavioral responses, capsaicin-activated currents, expression and membrane trafficking of PKC and TRPV1 in DRG. CPT was found to enhance capsaicin-induced nociception and ionic currents. The enhancement was inhibited by PKCα and PKCε inhibitors. In addition, CPT increased the expression of phosphorylated PKCα (pPKCα) and membrane TRPV1 expression in DRG. Studying the colocalization of TRPV1 and pPKCα or pPKCε in DRG slices prepared from CFA-treated rats, we found that pPKCα or pPKCε expressed with TRPV1 in different-sized neurons to exert differential influences on TRPV1 activity. Thus, Epac-PKC signaling is critically important in producing inflammation-induced potentiation of TRPV1 functions.


Subject(s)
Acetylcysteine/analogs & derivatives , Erythromycin/analogs & derivatives , Hyperalgesia/physiopathology , Inflammation/enzymology , Protein Kinase C-epsilon/metabolism , Signal Transduction/physiology , TRPV Cation Channels/metabolism , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biotinylation , Capsaicin/toxicity , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Erythromycin/metabolism , Erythromycin/pharmacology , Freund's Adjuvant/toxicity , Ganglia, Spinal/cytology , Hyperalgesia/pathology , Inflammation/chemically induced , Male , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Protein Kinase C-alpha/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4/metabolism
3.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27385722

ABSTRACT

Sensitization of purinergic P2X3 receptors (P2X3Rs) contributes to the production of exaggerated nociceptive responses following inflammatory injury. We showed previously that prostaglandin E2 (PGE2) potentiates P2X3R-mediated ATP currents in dorsal root ganglion neurons isolated from both control and complete Freund's adjuvant-induced inflamed rats. PGE2 potentiation of ATP currents depends only on PKA signaling in control neurons, but it depends on both PKA and PKC signaling in inflamed neurons. We further found that inflammation evokes an increase in exchange proteins directly activated by cAMP (Epacs) in dorsal root ganglions. This increase promotes the activation of PKC to produce a much enhanced PGE2 effect on ATP currents and to elicit Epac-dependent flinch nocifensive behavioral responses in complete Freund's adjuvant rats. The link between Epac-PKC signaling and P2X3R sensitization remains unexplored. Here, we show that the activation of Epacs promotes the expression of phosphorylated PKC and leads to an increase in the cytoskeleton, F-actin, expression at the cell perimeter. Depolymerization of F-actin blocks PGE2-enhanced ATP currents and inhibits P2X3R-mediated nocifensive responses after inflammation. Thus, F-actin is dynamically involved in the Epac-PKC-dependent P2X3R sensitization. Furthermore, Epacs induce a PKC-dependent increase in the membrane expression of P2X3Rs. This increase is abolished by F-actin depolymerization, suggesting that F-actin mediates Epac-PKC signaling of P2X3R membrane expression. Thus, after inflammation, an Epac-PKC dependent increase in F-actin in dorsal root ganglion neurons enhances the membrane expression of P2X3Rs to bring about sensitization of P2X3Rs and abnormal pain behaviors.


Subject(s)
Actins/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Guanine Nucleotide Exchange Factors/metabolism , Inflammation/pathology , Protein Kinase C/metabolism , Receptors, Purinergic P2X3/metabolism , Signal Transduction , Adenosine Triphosphate/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cytochalasin D/pharmacology , Dinoprostone/pharmacology , Freund's Adjuvant , Ganglia, Spinal/drug effects , Hyperalgesia/pathology , Inflammation/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Thionucleotides/pharmacology
4.
Pain ; 157(7): 1541-1550, 2016 07.
Article in English | MEDLINE | ID: mdl-26963850

ABSTRACT

Sensitization of purinergic P2X3 receptors (P2X3Rs) is a major mechanism contributing to injury-induced exaggerated pain responses. We showed in a previous study that cyclic adenosine monophosphate (cAMP)-dependent guanine nucleotide exchange factor 1 (Epac1) in rat sensory dorsal root ganglia (DRGs) is upregulated after inflammatory injury, and it plays a critical role in P2X3R sensitization by activating protein kinase C epsilon (PKCε) inside the cells. protein kinase C epsilon has been established as the major PKC isoform mediating injury-induced hyperalgesic responses. On the other hand, the role of PKCα in receptor sensitization was seldom considered. Here, we studied the participation of PKCα in Epac signaling in P2X3R-mediated hyperalgesia. The expression of both Epac1 and Epac2 and the level of cAMP in DRGs are greatly enhanced after complete Freund adjuvant (CFA)-induced inflammation. The expression of phosphorylated PKCα is also upregulated. Complete Freund adjuvant (CFA)-induced P2X3R-mediated hyperalgesia is not only blocked by Epac antagonists but also by the classical PKC isoform inhibitors, Go6976, and PKCα-siRNA. These CFA effects are mimicked by the application of the Epac agonist, 8-(4-chlorophenylthio)-2 -O-methyl-cAMP (CPT), in control rats, further confirming the involvement of Epacs. Because the application of Go6976 prior to CPT still reduces CPT-induced hyperalgesia, PKCα is downstream of Epacs to mediate the enhancement of P2X3R responses in DRGs. The pattern of translocation of PKCα inside DRG neurons in response to CPT or CFA stimulation is distinct from that of PKCε. Thus, in contrast to prevalent view, PKCα also plays an essential role in producing complex inflammation-induced receptor-mediated hyperalgesia.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Protein Kinase C-alpha/metabolism , Receptors, Purinergic P2X3/metabolism , Signal Transduction/physiology , Animals , Carbazoles/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Male , Protein Kinase C-alpha/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Mol Pain ; 11: 68, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26542462

ABSTRACT

BACKGROUND: We have previously shown that endogenously active purinergic P2X7 receptors (P2X7Rs) in satellite glial cells of dorsal root ganglia (DRGs) stimulate ATP release. The ATP activates P2Y1Rs located in the enwrapped neuronal somata, resulting in down-regulation of P2X3Rs. This P2X7R-P2Y1-P2X3R inhibitory control significantly reduces P2X3R-mediated nociceptive responses. The underlying mechanism by which the activation of P2Y1Rs inhibits the expression of P2X3Rs remains unexplored. RESULTS: Examining the effect of the activation of p38 mitogen-activated protein kinase on the expression of P2X3Rs in DRGs, we found that the p38 activator, anisomycin (Anis), reduced the expression of P2X3Rs. Blocking the activity of SGCs by the glial Krebs cycle inhibitor, fluorocitrate, did not change the effect of Anis. These results suggest that neuronal p38 plays a major role in the inhibition of P2X3R expression. Western blotting analyses showed that inhibiting P2Y1Rs by MRS2179 (MRS) or blocking P2X7Rs by either oxATP or A740003 reduced pp38 and increased P2X3R expression in DRGs. These results are further supported by the immunohistochemical study showing that P2X7R and P2Y1R antagonists reduce the percentage of pp38-positive neurons. These observations suggest that activation of P2X7Rs and P2Y1Rs promotes p38 activity to exert inhibitory control on P2X3R expression. Since activation of p38 by Anis in the presence of either A740003 or MRS could overcome the block of P2X7R-P2Y1R inhibitory control, p38 in DRG neurons is downstream of P2Y1Rs. In addition, inhibition of p38 by SB202190 was found to prevent the P2X7R and P2Y1R block of P2X3R expression and increase P2X3R-mediated nociceptive flinch behaviors. CONCLUSIONS: p38 in DRG neurons downstream of P2Y1R is necessary and sufficient for the P2X7R-P2Y1R inhibitory control of P2X3R expression.


Subject(s)
Ganglia, Spinal/physiology , Receptors, Purinergic P2X3/genetics , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2Y1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Down-Regulation/drug effects , Enzyme Activation/drug effects , Ganglia, Spinal/metabolism , Imidazoles/pharmacology , Male , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
6.
Mol Pain ; 9: 63, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24304623

ABSTRACT

BACKGROUND: Phosphorylation sites in the C-terminus of mu-opioid receptors (MORs) are known to play critical roles in the receptor functions. Our understanding of their participation in opioid analgesia is mostly based on studies of opioid effects on mutant receptors expressed in in vitro preparations, including cell lines, isolated neurons and brain slices. The behavioral consequences of the mutation have not been fully explored due to the complexity in studies of mutant receptors in vivo. To facilitate the determination of the contribution of phosphorylation sites in MOR to opioid-induced analgesic behaviors, we expressed mutant and wild-type human MORs (hMORs) in sensory dorsal root ganglion (DRG) neurons, a major site for nociceptive (pain) signaling and determined morphine- and the full MOR agonist, DAMGO,-induced effects on heat-induced hyperalgesic behaviors and potassium current (IK) desensitization in these rats. FINDINGS: A mutant hMOR DNA with the putative phosphorylation threonine site at position 394 replaced by an alanine (T394A), i.e., hMOR-T, or a plasmid containing wild type hMOR (as a positive control) was intrathecally delivered. The plasmid containing GFP or saline was used as the negative control. To limit the expression of exogenous DNA to neurons of DRGs, a neuron-specific promoter was included in the plasmid. Following a plasmid injection, hMOR-T or hMOR receptors were expressed in small and medium DRG neurons. Compared with saline or GFP rats, the analgesic potency of morphine was increased to a similar extent in hMOR-T and hMOR rats. Morphine induced minimum IK desensitization in both rat groups. In contrast, DAMGO increased analgesic potency and elicited IK desensitization to a significantly less extent in hMOR-T than in hMOR rats. The development and extent of acute and chronic tolerance induced by repeated morphine or DAMGO applications were not altered by the T394A mutation. CONCLUSIONS: These results indicate that phosphorylation of T394 plays a critical role in determining the potency of DAMGO-induced analgesia and IK desensitization, but has limited effect on morphine-induced responses. On the other hand, the mutation contributes minimally to both DAMGO- and morphine-induced behavioral tolerance. Furthermore, the study shows that plasmid gene delivery of mutant receptors to DRG neurons is a useful strategy to explore nociceptive behavioral consequences of the mutation.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/genetics
7.
J Neurosci ; 33(21): 9028-38, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23699514

ABSTRACT

Patients with long-standing diabetes frequently demonstrate gastric hypersensitivity with an unknown mechanism. The present study was designed to investigate roles for nuclear factor-κB (NF-κB) and the endogenous H2S-producing enzyme cystathionine-ß-synthetase (CBS) signaling pathways by examining cbs gene methylation status in adult rats with diabetes. Intraperitoneal injection of streptozotocin (STZ) produced gastric hypersensitivity in female rats in response to gastric balloon distention. Treatment with the CBS inhibitor aminooxyacetic acid significantly attenuated STZ-induced gastric hypersensitivity in a dose-dependent fashion. Aminooxyacetic acid treatment also reversed hyperexcitability of gastric-specific dorsal root ganglion (DRG) neurons labeled by the dye DiI in diabetic rats. Conversely, the H2S donor NaHS enhanced neuronal excitability of gastric DRG neurons. Expression of CBS and p65 were markedly enhanced in gastric DRGs in diabetic rats. Blockade of NF-κB signaling using pyrrolidine dithiocarbamate reversed the upregulation of CBS expression. Interestingly, STZ treatment led to a significant demethylation of CpG islands in the cbs gene promoter region, as determined by methylation-specific PCR and bisulfite sequencing. STZ treatment also remarkably downregulated the expression of DNA methyltransferase 3a and 3b. More importantly, STZ treatment significantly enhanced the ability of cbs to bind DNA at the p65 consensus site, as shown by chromatin immunoprecipitation assays. Our findings suggest that upregulation of cbs expression is attributed to cbs promoter DNA demethylation and p65 activation and that the enhanced interaction of the cbs gene and p65 contributes to gastric hypersensitivity in diabetes. This finding may guide the development and evaluation of new treatment modalities for patients with diabetic gastric hypersensitivity.


Subject(s)
Cystathionine beta-Synthase/metabolism , Diabetes Mellitus, Experimental/complications , Hypersensitivity , NF-kappa B/metabolism , Stomach Diseases/etiology , Amino Acids , Analysis of Variance , Animals , Area Under Curve , Case-Control Studies , Chromatin Immunoprecipitation , CpG Islands/drug effects , CpG Islands/physiology , Cystathionine beta-Synthase/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Disease Models, Animal , Dose-Response Relationship, Drug , Electromyography , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/pathology , Hypersensitivity/drug therapy , Hypersensitivity/etiology , Membrane Potentials/drug effects , Methylation/drug effects , Neoplasm Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Oxamic Acid/therapeutic use , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Stomach Diseases/drug therapy , Sulfites/pharmacology , Up-Regulation/drug effects , DNA Methyltransferase 3B
8.
J Pain Res ; 5: 415-24, 2012.
Article in English | MEDLINE | ID: mdl-23166445

ABSTRACT

BACKGROUND: Evaluation of analgesics in large animals is a necessary step in the development of better pain medications or gene therapy prior to clinical trials. However, chronic neuropathic pain models in large animals are limited. To address this deficiency, we developed a neuropathic pain model in sheep, which shares many anatomical similarities in spine dimensions and cerebrospinal fluid volume as humans. METHODS: A neuropathic pain state was induced in sheep by tight ligation and axotomy of the common peroneal nerve. The analgesic effect of intrathecal (IT) morphine was investigated. Interspecies comparison was conducted by analyzing the ceiling doses of IT morphine for humans, sheep, and rats. RESULTS: Peroneal nerve injury (PNI) produced an 86% decrease in von-Frey filament-evoked withdrawal threshold on postsurgery day 3 and the decrease lasted for the 8-week test period. Compared to the pre-injury, sham, and contralateral hindlimb, the IT morphine dose that produces 50% of maximum analgesia (ED(50)) for injured PNI hindlimb was 1.8-fold larger and E(max), the dose that produces maximal analgesia, was 6.1-fold lower. The sheep model closely predicts human IT morphine ceiling dose by allometric scaling. This is in contrast to the approximately 10-fold lower morphine ceiling dose predicted by the rat spinal nerve ligated or spared nerve injury models. CONCLUSION: PNI sheep model has a fast onset and shows stable and long-lasting pain behavioral characteristics. Since the antinociceptive properties of IT morphine are similar to those observed in humans, the PNI sheep model will be a useful tool for the development of analgesics. Its large size and consistent chronic pain behavior will facilitate the development and evaluation of surgical intervention and gene therapy. The PNI sheep pain model provides us with the opportunity for multi-species testing, which will improve the success of clinical trials.

9.
Mol Pain ; 8: 9, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22314033

ABSTRACT

BACKGROUND: The purinergic P2X3 receptor (P2X3R) expressed in the dorsal root ganglion (DRG) sensory neuron and the P2X7 receptor (P2X7R) expressed in the surrounding satellite glial cell (SGC) are two major receptors participating in neuron-SGC communication in adult DRGs. Activation of P2X7Rs was found to tonically reduce the expression of P2X3Rs in DRGs, thus inhibiting the abnormal pain behaviors in adult rats. P2X receptors are also actively involved in sensory signaling in developing rodents. However, very little is known about the developmental change of P2X7Rs in DRGs and the interaction between P2X7Rs and P2X3Rs in those animals. We therefore examined the expression of P2X3Rs and P2X7Rs in postnatal rats and determined if P2X7R-P2X3R control exists in developing rats. FINDINGS: We immunostained DRGs of immature rats and found that P2X3Rs were expressed only in neurons and P2X7Rs were expressed only in SGCs. Western blot analyses indicated that P2X3R expression decreased while P2X7R expression increased with the age of rats. Electrophysiological studies showed that the number of DRG neurons responding to the stimulation of the P2XR agonist, α,ß-meATP, was higher and the amplitudes of α,ß-meATP-induced depolarizations were larger in immature DRG neurons. As a result, P2X3R-mediated flinching responses were much more pronounced in immature rats than those found in adult rats. When we reduced P2X7R expression with P2X7R-siRNA in postnatal and adult rats, P2X3R-mediated flinch responses were greatly enhanced in both rat populations. CONCLUSIONS: These results show that the P2X7R expression increases as rats age. In addition, P2X7Rs in SGCs exert inhibitory control on the P2X3R expression and function in sensory neurons of immature rats, just as observed in adult rats. Regulation of P2X7R expression is likely an effective way to control P2X3R activity and manage pain relief in infants.


Subject(s)
Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Neurons/metabolism , Receptors, Purinergic P2X3/metabolism , Receptors, Purinergic P2X7/metabolism , Satellite Cells, Perineuronal/metabolism , Aging/metabolism , Animals , Ganglia, Spinal/cytology , Rats , Rats, Sprague-Dawley , Satellite Cells, Perineuronal/cytology
10.
Mol Pain ; 7: 60, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21851615

ABSTRACT

BACKGROUND: Diabetic neuropathy is a common neuropathy associated with paresthaesia and pain. The mechanisms underlying the painful conditions are not well understood. The aim of this study is to investigate the participation of purinergic P2X3 receptors in painful diabetic neuropathy. RESULTS: Diabetes was induced by an intraperitoneal injection of streptozotocin (STZ). We showed that mechanical allodynia was induced two weeks after a STZ injection and lasted for at least another seven weeks. The mechanical allodynia was significantly attenuated by peripheral administration of the P2X receptor antagonists, PPADS or TNP-ATP. DiI was subcutaneously injected into the rat hindpaw to label hindpaw-innervated dorsal root ganglion (DRG) neurons. ATP activated fast-inactivating P2X3 receptor-mediated currents in the labeled DRG neurons were studied. ATP responses in STZ-treated rats were ~2-fold larger than those in control rats. Furthermore, the expression of P2X3 receptor proteins in the plasma membrane of L4-6 DRGs of STZ rats was significantly enhanced while the total expression of P2X3 receptors remained unaltered. CONCLUSIONS: These results indicate that a large enhancement of P2X3 receptor activity and an increase in the membrane expression of P2X3 receptors contribute to the development of chronic pain in STZ-induced diabetic rats and suggest a possible target for the treatment of diabetic neuropathic pain.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Hyperalgesia/complications , Hyperalgesia/metabolism , Receptors, Purinergic P2X3/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Diabetes Mellitus, Experimental/pathology , Extremities/innervation , Extremities/pathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Hyperalgesia/pathology , Hyperglycemia/complications , Hyperglycemia/metabolism , Hyperglycemia/pathology , Ion Channel Gating/drug effects , Male , Purinergic P2X Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Streptozocin , Time Factors , Up-Regulation/drug effects
11.
Neuron Glia Biol ; 6(1): 53-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20604979

ABSTRACT

It has been known for some time that the somata of neurons in sensory ganglia respond to electrical or chemical stimulation and release transmitters in a Ca2+-dependent manner. The function of the somatic release has not been well delineated. A unique characteristic of the ganglia is that each neuronal soma is tightly enwrapped by satellite glial cells (SGCs). The somatic membrane of a sensory neuron rarely makes synaptic contact with another neuron. As a result, the influence of somatic release on the activity of adjacent neurons is likely to be indirect and/or slow. Recent studies of neuron-SGC interactions have demonstrated that ATP released from the somata of dorsal root ganglion neurons activates SGCs. They in turn exert complex excitatory and inhibitory modulation of neuronal activity. Thus, SGCs are actively involved in the processing of afferent information. In this review, we summarize our understanding of bidirectional communication between neuronal somata and SGCs in sensory ganglia and its possible role in afferent signaling under normal and injurious conditions. The participation of purinergic receptors is emphasized because of their dominant roles in the communication.


Subject(s)
Cell Communication/physiology , Ganglia, Sensory/cytology , Neuroglia/physiology , Neurons/physiology , Receptors, Purinergic/physiology , Adenosine Triphosphate/metabolism , Animals , Biofeedback, Psychology/physiology , Models, Biological , Neurons/cytology , RNA, Messenger/metabolism , Receptors, Purinergic/classification , Receptors, Purinergic/genetics
12.
Proc Natl Acad Sci U S A ; 105(43): 16773-8, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18946042

ABSTRACT

Purinergic ionotropic P2X7 receptors (P2X7Rs) are closely associated with excitotoxicity and nociception. Inhibition of P2X7R activation has been considered as a potentially useful strategy to improve recovery from spinal cord injury and reduce inflammatory damage to trauma. The physiological functions of P2X7Rs, however, are poorly understood, even though such information is essential for making the P2X7R an effective therapeutic target. We show here that P2X7Rs in satellite cells of dorsal root ganglia tonically inhibit the expression of P2X3Rs in neurons. Reducing P2X7R expression using siRNA or blocking P2X7R activity by antagonists elicits P2X3R up-regulation, increases the activity of sensory neurons responding to painful stimuli, and evokes abnormal nociceptive behaviors in rats. Thus, contrary to the notion that P2X7R activation is cytotoxic, P2X7Rs in satellite cells play a crucial role in maintaining proper P2X3R expression in dorsal root ganglia. Studying the mechanism underlying the P2X7R-P2X3R control, we demonstrate that activation of P2X7Rs evokes ATP release from satellite cells. ATP in turn stimulates P2Y1 receptors in neurons. P2Y1 receptor activation appears to be necessary and sufficient for the inhibitory control of P2X3R expression. We further determine the roles of the P2X7R-P2Y1-P2X3R inhibitory control under injurious conditions. Activation of the inhibitory control effectively prevents the development of allodynia and increases the potency of systemically administered P2X7R agonists in inflamed rats. Thus, direct blocking P2X7Rs, as proposed before, may not be the best strategy for reducing pain or lessening neuronal degeneration because it also disrupts the protective function of P2X7Rs.


Subject(s)
Down-Regulation/genetics , Neuroglia/metabolism , Pain , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/physiology , Satellite Cells, Perineuronal/metabolism , Animals , Ganglia, Spinal , Male , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3 , Receptors, Purinergic P2X7 , Sensory Receptor Cells/chemistry
13.
J Physiol ; 584(Pt 1): 191-203, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17702820

ABSTRACT

Sensitization of purinergic P2X receptors is one of the mechanisms responsible for exaggerated pain responses to inflammatory injuries. Prostaglandin E2 (PGE2), produced by inflamed tissues, is known to contribute to abnormal pain states. In a previous study, we showed that PGE2 increases fast inactivating ATP currents that are mediated by homomeric P2X3 receptors in dorsal root ganglion (DRG) neurons isolated from normal rats. Protein kinase A (PKA) is the signalling pathway used by PGE2. Little is known about the action of PGE2 on ATP currents after inflammation, although the information is crucial for understanding the mechanisms underlying inflammation-induced sensitization of P2X receptors. We therefore studied the effects of PGE2 on P2X3 receptor-mediated ATP currents in DRG neurons dissociated from complete Freund's adjuvant (CFA)-induced inflamed rats. We found that PGE2 produces a large increase in ATP currents. PKCepsilon, in addition to PKA, becomes involved in the modulatory action of PGE2. Thus, PGE2 signalling switches from a solely PKA-dependent pathway under normal conditions to both PKA- and PKC-dependent pathways after inflammation. Studying the mechanisms underlying the switch, we demonstrated that cAMP-responsive guanine nucleotide exchange factor 1 (Epac1) is up-regulated after inflammation. The Epac agonist CPT-OMe mimics the potentiating effect of PGE2 and occludes the PKC-mediated PGE2 action on ATP currents. These results suggest that Epac plays a critical role in P2X3 sensitization by activation of de novo PKC-dependent signalling of PGE2 after inflammation and would be a useful therapeutic target for pain therapies.


Subject(s)
Dinoprostone/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Inflammation/metabolism , Nociceptors/metabolism , Protein Kinase C-epsilon/metabolism , Adenosine Triphosphate/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Ganglia, Spinal/metabolism , Rats , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3 , Signal Transduction/physiology
14.
Mol Pain ; 3: 22, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17692121

ABSTRACT

Prostaglandin E2 (PGE2) is a well-known inflammatory mediator that enhances the excitability of DRG neurons. Homomeric P2X3 and heteromeric P2X2/3 receptors are abundantly expressed in dorsal root ganglia (DRG) neurons and participate in the transmission of nociceptive signals. The interaction between PGE2 and P2X3 receptors has not been well delineated. We studied the actions of PGE2 on ATP-activated currents in dissociated DRG neurons under voltage-clamp conditions. PGE2 had no effects on P2X2/3 receptor-mediated responses, but significantly potentiated fast-inactivating ATP currents mediated by homomeric P2X3 receptors. PGE2 exerted its action by activating EP3 receptors. To study the mechanism underlying the action of PGE2, we found that the adenylyl cyclase activator, forskolin and the membrane-permeable cAMP analogue, 8-Br-cAMP increased ATP currents, mimicking the effect of PGE2. In addition, forskolin occluded the enhancement produced by PGE2. The protein kinase A (PKA) inhibitors, H89 and PKA-I blocked the PGE2 effect. In contrast, the PKC inhibitor, bisindolymaleimide (Bis) did not change the potentiating action of PGE2. We further showed that PGE2 enhanced alpha,beta-meATP-induced allodynia and hyperalgesia and the enhancement was blocked by H89. These observations suggest that PGE2 binds to EP3 receptors, resulting in the activation of cAMP/PKA signaling pathway and leading to an enhancement of P2X3 homomeric receptor-mediated ATP responses in DRG neurons.


Subject(s)
Dinoprostone/pharmacology , Ganglia, Spinal/drug effects , Neurons/drug effects , Purinergic P2 Receptor Agonists , Adenosine Triphosphate/metabolism , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X3
15.
Pain ; 119(1-3): 38-48, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16298067

ABSTRACT

P2X3 and P2X2/3 receptors in dorsal root ganglia (DRG) appear to participate in producing nociceptive responses after nerve injury. However, the mechanisms underlying the receptor-mediated nociception in the neuropathic state remain unclear. Using spared nerve injury (SNI) rats, we found that allodynic and nocifensive (flinch) behavioral responses developed after injury can be reversed by P2X receptor antagonists, indicating an involvement of P2X receptors. Immunocytochemical studies revealed that P2X3 receptors are expressed in small and medium but rarely in large DRG neurons of both normal and SNI rats. Thus, contrary to the conventional view that only large A beta cells mediate allodynia, small and medium cells are intimately involved in P2X3 receptor-mediated allodynia. Measuring ATP levels in the subcutaneous space of the rat paw, we showed that ATP release does not change after SNI. On the other hand, the P2X receptor agonist, alpha beta-methylene ATP produces 3.5-fold larger flinch responses at a 8.0-fold lower dose. Thus, sensitization of P2X3 receptors rather than a change in ATP release is responsible for the neuropathic pain behaviors. We further demonstrated that sensitization of P2X3 receptors arises from an increase in receptor function. ATP-induced P2X3 receptor-mediated currents in DRG neurons is 2.5-fold larger after SNI. The expression of P2X3 receptors on the cell membrane is significantly enhanced while the total expression of P2X3 receptors remained unchanged. Thus, the enhancement of trafficking of P2X3 receptors is likely an important mechanism contributing to the increase in receptor function after nerve injury.


Subject(s)
Ganglia, Spinal/metabolism , Hyperesthesia/metabolism , Neuralgia/metabolism , Neurons/metabolism , Receptors, Purinergic P2/metabolism , Sciatic Nerve/injuries , Animals , Hyperesthesia/etiology , Male , Neuralgia/complications , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X3
16.
J Pain ; 6(7): 447-54, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15993823

ABSTRACT

UNLABELLED: We have shown previously that using recombinant adeno-associated viral vector (rAAV) to up-regulate mu opioid receptors (muORs) in dorsal root ganglia (DRGs) increases the potency of subcutaneous morphine. Here we report an improved method of introducing rAAV-muOR viral vectors into DRGs. Instead of injecting the rAAV-muOR gene directly into DRGs as shown before, the vector was introduced into the sciatic nerve of rats. Changes in muOR expression and antinociceptive effects of intrathecal morphine in rAAV-muOR rats were examined. Immunocytochemical studies showed that the transduced muORs were expressed in all types (ie, small, medium, and large) of DRG neurons. The expression of muORs in DRG neurons, quantified by Western blotting, was increased by 1.7-fold 4 weeks after the sciatic nerve injection. The up-regulation persisted for more than 6 months. The effects of intrathecal morphine on paw withdrawal latencies to heat were studied in rats inflamed with complete Freund's adjuvant. Compared with rats injected with rAAV containing the enhanced green fluorescent protein gene (rAAV-EGFP), the antinociceptive potency of intrathecal morphine in rAAV-muOR rats was significantly increased, and the effective dose (ED50) for morphine was 5.4-fold lower (rAAV-muOR: ED50 = 0.84 microg, confidence interval, 0.70-0.99 microg; rAAV-EGFP: ED50 = 4.50 microg, confidence interval, 3.55-5.86 microg). With minimum tissue damage and a large persistent increase in the opioid potency, remote nerve injection of rAAV-muOR to up-regulate muORs could be a useful therapeutic strategy for the treatment of chronic pain. PERSPECTIVE: Injection of adeno-associated viral vector containing the muOR gene into the sciatic nerve produces a significant up-regulation of muORs in DRGs for up to 6 months without producing any immune responses in the injected animals. This results in a 5.4-fold increase in the potency of intrathecal morphine.


Subject(s)
Ganglia, Spinal/metabolism , Genetic Vectors/physiology , Morphine/pharmacology , Neurons, Afferent/metabolism , Receptors, Opioid, mu/genetics , Sciatic Nerve/metabolism , Transfection/methods , Analgesics, Opioid/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Genetic Therapy/methods , Genetic Therapy/trends , Injections, Spinal , Male , Microinjections/methods , Neurons, Afferent/drug effects , Nociceptors/drug effects , Nociceptors/metabolism , Pain Threshold/drug effects , Pain Threshold/physiology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Receptors, Opioid, mu/biosynthesis , Sciatic Nerve/cytology , Sciatic Nerve/surgery , Transduction, Genetic/methods , Up-Regulation/genetics
17.
Proc Natl Acad Sci U S A ; 101(32): 11868-73, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15292517

ABSTRACT

To elucidate the functional link between Ca(2+)/calmodulin protein kinase II (CaMKII) and P2X receptor activation, we studied the effects of electrical stimulation, such as occurs in injurious conditions, on P2X receptor-mediated ATP responses in primary sensory dorsal root ganglion neurons. We found that endogenously active CaMKII up-regulates basal P2X3 receptor activity in dorsal root ganglion neurons. Electrical stimulation causes prolonged increases in ATP currents that lasts up to approximately 45 min. In addition, the total and phosphorylated CaMKII are also up-regulated. The enhancement of ATP currents depends on Ca(2+) and calmodulin and is completely blocked by the CaMKII inhibitor, 2-[N-(2-hydroxyethyl)]-N-(4-methoxybenzenesulfonyl)]amino-N-(4-chlorocinnamyl)-N-methylbenzylamine). Western analyses indicate that electrical stimulation enhances the expression of P2X3 receptors in the membrane and that the enhancement is blocked by the inhibitor. These results suggest that CaMKII up-regulated by electrical stimulation enhances ATP responses by promoting trafficking of P2X receptors to the membrane and may play a key role in the sensitization of P2X receptors under injurious conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Receptors, Purinergic P2/metabolism , Animals , Calcium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cell Membrane/chemistry , Cell Membrane/metabolism , Electrophysiology , Kinetics , Neurons/metabolism , Phosphorylation , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/analysis , Receptors, Purinergic P2X3 , Up-Regulation/physiology
18.
Hum Gene Ther ; 14(9): 897-906, 2003 Jun 10.
Article in English | MEDLINE | ID: mdl-12828860

ABSTRACT

Transferring therapeutic genes into the nociceptive system, including dorsal root ganglia (DRGs) and the spinal cord, is potentially a powerful approach for the treatment of chronic pain in humans. Adeno-associated viral vectors (AAVs) are particularly useful in delivering foreign genes to targeted tissues because they seldom induce immune responses or produce cytotoxicity. To determine the efficiency of transgene expression and the best route(s) of delivery, a recombinant AAV type 2 vector containing the enhanced green fluorescent protein (EGFP) gene driven by the neuron-specific enolase (NSE) promoter (rAAV-EGFP) was constructed. We injected the vector into subcutaneous tissue, sciatic nerve, DRGs, and subarachnoid space, and examined EGFP expression in the DRG, spinal cord, and nerve fibers. Both sciatic nerve and DRG injection led to strong EGFP expression in a large number of DRG neurons. The expression persisted for more than 6-8 months. We then delivered the mu-opioid receptor (muOR) gene into DRGs through direct DRG or sciatic nerve injection of rAAV-muOR and found a significant increase in morphine efficacy. These results suggest that delivering therapeutic genes to DRGs by the rAAV-NSE vector is a valid strategy for treatment of chronic pain.


Subject(s)
Dependovirus/genetics , Ganglia, Spinal/metabolism , Genetic Vectors/administration & dosage , Receptors, Opioid, mu/genetics , Transgenes , Analgesics, Opioid/pharmacology , Animals , Behavior, Animal , Cells, Cultured , Dependovirus/immunology , Ganglia, Spinal/anatomy & histology , Ganglia, Spinal/cytology , Gene Expression , Green Fluorescent Proteins , Injections, Spinal , Injections, Subcutaneous , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Morphine/pharmacology , Nociceptors/metabolism , Posterior Horn Cells/metabolism , Rats , Sciatic Nerve , Subarachnoid Space , Transduction, Genetic
19.
J Neurosci ; 22(1): 93-102, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756492

ABSTRACT

ATP-gated P2X receptors in nociceptive sensory neurons participate in transmission of pain signals from the periphery to the spinal cord. To determine the role of P2X receptors under injurious conditions, we examined ATP-evoked responses in dorsal root ganglion (DRG) neurons isolated from rats with peripheral inflammation, induced by injections of complete Freund's adjuvant (CFA) into the hindpaw. Application of ATP induced both fast- and slow-inactivating currents in control and inflamed neurons. CFA treatment had no effect on the affinity of ATP for its receptors or receptor phenotypes. On the other hand, inflammation caused a twofold to threefold increase in both ATP-activated currents, altered the voltage dependence of P2X receptors, and enhanced the expression of P2X2 and P2X3 receptors. The increase in ATP responses gave rise to large depolarizations that exceeded the threshold of action potentials in inflamed DRG neurons. Thus, P2X receptor upregulation could account for neuronal hypersensitivity and contribute to abnormal pain responses associated with inflammatory injuries. These results suggest that P2X receptors are useful targets for inflammatory pain therapy.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Ganglia, Spinal/metabolism , Inflammation/physiopathology , Neurons/metabolism , Receptors, Purinergic P2/metabolism , Action Potentials/drug effects , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Binding, Competitive , Blotting, Western , Cell Separation , Dose-Response Relationship, Drug , Freund's Adjuvant , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hindlimb/innervation , Hindlimb/physiopathology , In Vitro Techniques , Inflammation/chemically induced , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Patch-Clamp Techniques , Phenotype , Rats , Rats, Sprague-Dawley
20.
Pain ; 93(1): 85-92, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406342

ABSTRACT

Gabapentin (Neurontin) (GBP) is a widely prescribed analgesic used in treating pain patients with peripheral nerve injuries, diabetic neuropathy and cancer. To understand the mechanism of its action, we used the whole-cell patch recording technique to study the effects of GBP on N-methyl-D-aspartate (NMDA)-evoked currents in single dorsal horn neurons isolated from normal rats and from rats with inflammation induced by the injection of complete Freund adjuvant (CFA) to the hindpaw. We found that GBP enhanced NMDA currents in normal neurons only when protein kinase C (PKC) was added to these cells. The enhancement resulted from an increase in the affinity of glycine for NMDA receptors by GBP. In contrast, in neurons isolated from CFA-treated rats, GBP enhanced NMDA responses without any PKC treatment. Since endogenous PKC in inflamed tissue is elevated, these results suggest that GBP exerts its effects only on those cells affected by inflammatory injuries. Thus, the effects of GBP on NMDA receptors are plastic; they depend on the phosphorylation states of cells or receptors. These observations point to a new strategy for drug design. A chemical whose action depends on the state of cells would maximize its effectiveness while keeping its side-effects to a minimum.


Subject(s)
Acetates/pharmacology , Amines , Cyclohexanecarboxylic Acids , Excitatory Amino Acid Antagonists/pharmacology , Protein Kinase C/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , gamma-Aminobutyric Acid , Animals , Cell Separation , Electrophysiology , Excitatory Amino Acid Agonists/pharmacology , Gabapentin , Glycine/metabolism , Immunohistochemistry , Male , N-Methylaspartate/pharmacology , Patch-Clamp Techniques , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL
...